1
|
Zampieri D, Romano M, Fortuna S, Amata E, Dichiara M, Cosentino G, Marrazzo A, Mamolo MG. Design, Synthesis, and Cytotoxic Assessment of New Haloperidol Analogues as Potential Anticancer Compounds Targeting Sigma Receptors. Molecules 2024; 29:2697. [PMID: 38893570 PMCID: PMC11173765 DOI: 10.3390/molecules29112697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Sigma receptors (SRs), including SR1 and SR2 subtypes, have attracted increasing interest in recent years due to their involvement in a wide range of activities, including the modulation of opioid analgesia, neuroprotection, and potential anticancer activity. In this context, haloperidol (HAL), a commonly used antipsychotic drug, also possesses SR activity and cytotoxic effects. Herein, we describe the identification of novel SR ligands, obtained by a chemical hybridization approach. There wereendowed with pan-affinity for both SR subtypes and evaluated their potential anticancer activity against SH-SY5Y and HUH-7 cancer cell lines. Through a chemical hybridization approach, we identified novel compounds (4d, 4e, 4g, and 4j) with dual affinity for SR1 and SR2 receptors. These compounds were subjected to cytotoxicity testing using a resazurin assay. The results revealed potent cytotoxic effects against both cancer cell lines, with IC50 values comparable to HAL. Interestingly, the cytotoxic potency of the novel compounds resembled that of the SR1 antagonist HAL rather than the SR2 agonist siramesine (SRM), indicating the potential role of SR1 antagonism in their mechanism of action. The further exploration of their structure-activity relationships and their evaluation in additional cancer cell lines will elucidate their therapeutic potential and may pave the way for the development of novel anticancer agents that target SRs.
Collapse
Affiliation(s)
- Daniele Zampieri
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy;
| | - Maurizio Romano
- Department of Life Sciences, University of Trieste, Via Valerio 28, 34127 Trieste, Italy;
| | - Sara Fortuna
- Cresset-New Cambridge House, Bassingbourn Road, Litlington, Cambridge SG8 0SS, UK;
| | - Emanuele Amata
- Department of Drug and Health Sciences, University of Catania, Viale Doria 6, 95125 Catania, Italy; (E.A.); (M.D.); (G.C.); (A.M.)
| | - Maria Dichiara
- Department of Drug and Health Sciences, University of Catania, Viale Doria 6, 95125 Catania, Italy; (E.A.); (M.D.); (G.C.); (A.M.)
| | - Giuseppe Cosentino
- Department of Drug and Health Sciences, University of Catania, Viale Doria 6, 95125 Catania, Italy; (E.A.); (M.D.); (G.C.); (A.M.)
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, University of Catania, Viale Doria 6, 95125 Catania, Italy; (E.A.); (M.D.); (G.C.); (A.M.)
| | - Maria Grazia Mamolo
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy;
| |
Collapse
|
2
|
Barbaraci C, di Giacomo V, Maruca A, Patamia V, Rocca R, Dichiara M, Di Rienzo A, Cacciatore I, Cataldi A, Balaha M, Rapino M, Zagni C, Zampieri D, Pasquinucci L, Parenti C, Amata E, Rescifina A, Alcaro S, Marrazzo A. Discovery of first novel sigma/HDACi dual-ligands with a potent in vitro antiproliferative activity. Bioorg Chem 2023; 140:106794. [PMID: 37659146 DOI: 10.1016/j.bioorg.2023.106794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 09/04/2023]
Abstract
Designing and discovering compounds for dual-target inhibitors is challenging to synthesize new, safer, and more efficient drugs than single-target drugs, especially to treat multifactorial diseases such as cancer. The simultaneous regulation of multiple targets might represent an alternative synthetic approach to optimize patient compliance and tolerance, minimizing the risk of target-based drug resistance due to the modulation of a few targets. To this end, we conceived for the first time the design and synthesis of dual-ligands σR/HDACi to evaluate possible employment as innovative candidates to address this complex disease. Among all synthesized compounds screened for several tumoral cell lines, compound 6 (Kiσ1R = 38 ± 3.7; Kiσ2R = 2917 ± 769 and HDACs IC50 = 0.59 µM) is the most promising candidate as an antiproliferative agent with an IC50 of 0.9 µM on the HCT116 cell line and no significant toxicity to normal cells. Studies of molecular docking, which confirmed the affinity over σ1R and a pan-HDACs inhibitory behavior, support a possible balanced affinity and activity between both targets.
Collapse
Affiliation(s)
- Carla Barbaraci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Viviana di Giacomo
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Annalisa Maruca
- Net4science academic spinoff srl, Università degli Studi "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy
| | - Vincenzo Patamia
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Roberta Rocca
- Net4science academic spinoff srl, Università degli Studi "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy
| | - Maria Dichiara
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Annalisa Di Rienzo
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Ivana Cacciatore
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Amelia Cataldi
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Marwa Balaha
- Department of Pharmacy, University "G. d'Annunzio", Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafr El Sheikh 33516, Egypt
| | - Monica Rapino
- Genetic Molecular Institute of CNR, Unit of Chieti, "G. d' Annunzio" University, Via dei Vestini 31, 66100 Chieti-Pescara, Italy
| | - Chiara Zagni
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Daniele Zampieri
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy
| | - Lorella Pasquinucci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Carmela Parenti
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Antonio Rescifina
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Stefano Alcaro
- Net4science academic spinoff srl, Università degli Studi "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy; Dipartimento di Scienze della Salute, Università degli Studi "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Viale Europa, 88100, Catanzaro, Italy.
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy.
| |
Collapse
|
3
|
Huang Q, Zang X, Zhang Z, Yu H, Ding B, Li Z, Cheng S, Zhang X, Ali MRK, Qiu X, Lv Z. Study on endogenous inhibitors against PD-L1: cAMP as a potential candidate. Int J Biol Macromol 2023; 230:123266. [PMID: 36646351 DOI: 10.1016/j.ijbiomac.2023.123266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/10/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
The discovery of new anti-cancer drugs targeting the PD-1/PD-L1 pathway has been a research hotspot in recent years. In this study, biological affinity ultrafiltration (BAU), UPLC-HRMS, molecular dynamic (MD) simulations and molecular docking methods were applied to search for endogenous active compounds that can inhibit the binding of PD-L1 to PD-1. We screened dozens of potential cancer related endogenous compounds. Surprisingly, cyclic adenosine monophosphate (cAMP) was found to have a direct inhibitory effect on the PD-1/PD-L1 binding with an in vitro IC50 value of about 36.4 ± 9.3 μM determined by homogeneous time-resolved fluorescence (HTRF) assay. cAMP could recover the proliferation of Jurkat T cells co-cultured with DU-145 cells and may suppress PD-L1 expression of DU-145 cells. cAMP was demonstrated to bind and induce PD-L1 dimerization by FRET assay, and also predicted by MD simulations and molecular docking. The finding of cAMP as a potential inhibitor directly targeting the PD-1/PD-L1 interaction could advance our understanding of the activity of endogenous compounds regulating PD-L1.
Collapse
Affiliation(s)
- Qiuyang Huang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China
| | - Xiaoling Zang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China; Laboratory of Marine Drugs and Biological Products, Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong 266235, PR China.
| | - Zhiwei Zhang
- College of Physics, Qingdao University, Qingdao, Shandong 266071, PR China
| | - Hang Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China
| | - Baoyan Ding
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China
| | - Zhuangzhuang Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China
| | - Simin Cheng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China
| | - Xin Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China
| | - Mustafa R K Ali
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xue Qiu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China; Laboratory of Marine Drugs and Biological Products, Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong 266235, PR China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, PR China; Laboratory of Marine Drugs and Biological Products, Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong 266235, PR China.
| |
Collapse
|
4
|
Fallica AN, Ciaffaglione V, Modica MN, Pittalà V, Salerno L, Amata E, Marrazzo A, Romeo G, Intagliata S. Structure-activity relationships of mixed σ1R/σ2R ligands with antiproliferative and anticancer effects. Bioorg Med Chem 2022; 73:117032. [DOI: 10.1016/j.bmc.2022.117032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
|
5
|
Han W, He W, Song Y, Zhao J, Song Z, Shan Y, Hua W, Sun Y. Multifunctional platinum(IV) complex bearing HDAC inhibitor and biotin moiety exhibits prominent cytotoxicity and tumor-targeting ability. Dalton Trans 2022; 51:7343-7351. [PMID: 35466968 DOI: 10.1039/d2dt00090c] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite the wide clinical use of platinum drugs in cancer treatment, their severe side effects and lack of tumor selectivity seriously limit their further clinical application. To address the limitations of the current platinum drugs, herein a multifunctional platinum(IV) compound 1 containing a histone deacetylase (HDAC) inhibitor (4-phenylbutyric acid, 4-PBA) and a tumor-targeting group (biotin) has been designed and prepared. An in vitro cytotoxicity study indicated that compound 1 exhibits comparable or superior cytotoxicity to cisplatin against the tested cancer cell lines, but greatly reduced toxicity in human normal liver LO2 cells, implying the potential tumor-targeting ability of compound 1. Molecular docking results indicate that compound 1 can effectively interact with a biotin-specific receptor (streptavidin) through its biotin moiety, enabling potential tumor-targeting capability. Further studies indicated that compound 1's cytotoxicity stems from inducing DNA damage via the mitochondrial apoptotic pathway and inhibiting HDACs. Consequently, this compound can not only take advantage of the tumor selectively of biotin to improve its tumor-targeting ability but also strengthen its anticancer activity via simultaneously targeting DNA and HDACs.
Collapse
Affiliation(s)
- Weinan Han
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Weiyu He
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Yutong Song
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Jian Zhao
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research and Pharmaceutical Research Center, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P.R. China.,Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, P.R. China
| | - Zhiheng Song
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Yi Shan
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Wuyang Hua
- School of Food Engineering, Jilin Agricultural Science and Technology University, Jilin 132000, P.R. China
| | - Yanyan Sun
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| |
Collapse
|
6
|
Repurposing Antipsychotics for Cancer Treatment. Biomedicines 2021; 9:biomedicines9121785. [PMID: 34944601 PMCID: PMC8698939 DOI: 10.3390/biomedicines9121785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 11/25/2022] Open
Abstract
Cancer is a leading cause of death worldwide, with approximately 19 million new cases each year. Lately, several novel chemotherapeutic drugs have been introduced, efficiently inhibiting tumor growth and proliferation. However, developing a new drug is a time- and money-consuming process, requiring around 1 billion dollars and nearly ten years, with only a minority of the initially effective anti-cancer drugs experimentally finally being efficient in human clinical trials. Drug repurposing for cancer treatment is an optimal alternative as the safety of these drugs has been previously tested, and thus, in case of successful preclinical studies, can be introduced faster and with a lower cost into phase 3 clinical trials. Antipsychotic drugs are associated with anti-cancer properties and, lately, there has been an increasing interest in their role in cancer treatment. In the present review, we discussed in detail the in-vitro and in-vivo properties of the most common typical and atypical antipsychotics, along with their mechanism of action.
Collapse
|
7
|
Barbaraci C, Giurdanella G, Leotta CG, Longo A, Amata E, Dichiara M, Pasquinucci L, Turnaturi R, Prezzavento O, Cacciatore I, Zuccarello E, Lupo G, Pitari GM, Anfuso CD, Marrazzo A. Haloperidol Metabolite II Valproate Ester ( S)-(-)-MRJF22: Preliminary Studies as a Potential Multifunctional Agent Against Uveal Melanoma. J Med Chem 2021; 64:13622-13632. [PMID: 34477381 PMCID: PMC8474110 DOI: 10.1021/acs.jmedchem.1c00995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Increased angiogenesis and vascular endothelial growth factor (VEGF) levels contribute to higher metastasis and mortality in uveal melanoma (UM), an aggressive malignancy of the eye in adults. (±)-MRJF22, a prodrug of the sigma (σ) ligand haloperidol metabolite II conjugated with the histone deacetylase (HDAC) inhibitor valproic acid, has previously demonstrated a promising antiangiogenic activity. Herein, the asymmetric synthesis of (R)-(+)-MRJF22 and (S)-(-)-MRJF22 was performed to investigate their contribution to (±)-MRJF22 antiangiogenic effects in human retinal endothelial cells (HREC) and to assess their therapeutic potential in primary human uveal melanoma (UM) 92-1 cell line. While both enantiomers displayed almost identical capabilities to reduce cell viability than the racemic mixture, (S)-(-)-MRJF22 exhibited the highest antimigratory effects in endothelial and tumor cells. Given the fundamental contribution of cell motility to cancer progression, (S)-(-)-MRJF22 may represent a promising candidate for novel antimetastatic therapy in patients with UM.
Collapse
Affiliation(s)
- Carla Barbaraci
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.,Vera Salus Ricerca S.r.l., Via Sigmund Freud 62/B, 96100 Siracusa, Italy
| | - Giovanni Giurdanella
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | | | - Anna Longo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | - Emanuele Amata
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Maria Dichiara
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Lorella Pasquinucci
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Rita Turnaturi
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Ivana Cacciatore
- Department of Pharmacy, "G. D'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti Scalo, Italy
| | - Elisa Zuccarello
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York 10032, United States
| | - Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | | | - Carmelina Daniela Anfuso
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
8
|
Abatematteo FS, Niso M, Lacivita E, Abate C. σ 2 Receptor and Its Role in Cancer with Focus on a MultiTarget Directed Ligand (MTDL) Approach. Molecules 2021; 26:3743. [PMID: 34205334 PMCID: PMC8235595 DOI: 10.3390/molecules26123743] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/19/2022] Open
Abstract
Sigma-2 (σ2) is an endoplasmic receptor identified as the Endoplasmic Reticulum (ER) transmembrane protein TMEM97. Despite its controversial identity, which was only recently solved, this protein has gained scientific interest because of its role in the proliferative status of cells; many tumor cells from different organs overexpress the σ2 receptor, and many σ2 ligands display cytotoxic actions in (resistant) cancer cells. These properties have shed light on the σ2 receptor as a potential druggable target to be bound/activated for the diagnosis or therapy of tumors. Additionally, diverse groups have shown how the σ2 receptor can be exploited for the targeted delivery of the anticancer drugs to tumors. As the cancer disease is a multifactorial pathology with multiple cell populations, a polypharmacological approach is very often needed. Instead of the simultaneous administration of different classes of drugs, the use of one molecule that interacts with diverse pharmacological targets, namely MultiTarget Directed Ligand (MTDL), is a promising and currently pursued strategy, that may overcome the pharmacokinetic problems associated with the administration of multiple molecules. This review aims to point out the progress regarding the σ2 ligands in the oncology field, with a focus on MTDLs directed towards σ2 receptors as promising weapons against (resistant) cancer diseases.
Collapse
Affiliation(s)
| | | | | | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy; (F.S.A.); (M.N.); (E.L.)
| |
Collapse
|
9
|
Abatematteo FS, Niso M, Contino M, Leopoldo M, Abate C. Multi-Target Directed Ligands (MTDLs) Binding the σ 1 Receptor as Promising Therapeutics: State of the Art and Perspectives. Int J Mol Sci 2021; 22:6359. [PMID: 34198620 PMCID: PMC8232171 DOI: 10.3390/ijms22126359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/31/2021] [Accepted: 06/10/2021] [Indexed: 11/18/2022] Open
Abstract
The sigma-1 (σ1) receptor is a 'pluripotent chaperone' protein mainly expressed at the mitochondria-endoplasmic reticulum membrane interfaces where it interacts with several client proteins. This feature renders the σ1 receptor an ideal target for the development of multifunctional ligands, whose benefits are now recognized because several pathologies are multifactorial. Indeed, the current therapeutic regimens are based on the administration of different classes of drugs in order to counteract the diverse unbalanced physiological pathways associated with the pathology. Thus, the multi-targeted directed ligand (MTDL) approach, with one molecule that exerts poly-pharmacological actions, may be a winning strategy that overcomes the pharmacokinetic issues linked to the administration of diverse drugs. This review aims to point out the progress in the development of MTDLs directed toward σ1 receptors for the treatment of central nervous system (CNS) and cancer diseases, with a focus on the perspectives that are proper for this strategy. The evidence that some drugs in clinical use unintentionally bind the σ1 protein (as off-target) provides a proof of concept of the potential of this strategy, and it strongly supports the promise that the σ1 receptor holds as a target to be hit in the context of MTDLs for the therapy of multifactorial pathologies.
Collapse
Affiliation(s)
| | | | | | | | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy; (F.S.A.); (M.N.); (M.C.); (M.L.)
| |
Collapse
|
10
|
Oyer HM, Sanders CM, Kim FJ. Small-Molecule Modulators of Sigma1 and Sigma2/TMEM97 in the Context of Cancer: Foundational Concepts and Emerging Themes. Front Pharmacol 2019; 10:1141. [PMID: 31695608 PMCID: PMC6816035 DOI: 10.3389/fphar.2019.01141] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/04/2019] [Indexed: 12/17/2022] Open
Abstract
There are two known subtypes of the so-called sigma receptors, Sigma1 and Sigma2. Sigma1 (encoded by the SIGMAR1 gene and also known as Sigma-1 receptor, S1R) is a unique pharmacologically regulated integral membrane chaperone or scaffolding protein that allosterically modulates the activity of its associated proteins. Sigma2, recently identified as transmembrane protein 97 (TMEM97), is an integral membrane protein implicated in cellular cholesterol homeostasis. A number of publications over the past two decades have suggested a role for both sigma proteins in tumor biology. Although there is currently no clinically used anti-cancer drug that targets Sigma1 or Sigma2/TMEM97, a growing body of evidence supports the potential of small-molecule compounds with affinity for these proteins, putative sigma ligands, as therapeutic agents to treat cancer. In preclinical models, these compounds have been reported to inhibit cancer cell proliferation, survival, adhesion, and migration; furthermore, they have been demonstrated to suppress tumor growth, to alleviate cancer-associated pain, and to exert immunomodulatory properties. Here, we will address the known knowns and the known unknowns of Sigma1 and Sigma2/TMEM97 ligand actions in the context of cancer. This review will highlight key discoveries and published evidence in support of a role for sigma proteins in cancer and will discuss several fundamental questions regarding the physiological roles of sigma proteins in cancer and sigma ligand mechanism of action.
Collapse
Affiliation(s)
- Halley M Oyer
- Department of Cancer Biology, Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, PA, United States
| | - Christina M Sanders
- Department of Cancer Biology, Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, PA, United States
| | - Felix J Kim
- Department of Cancer Biology, Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
11
|
Molecular mechanisms of anti-psychotic drugs for improvement of cancer treatment. Eur J Pharmacol 2019; 856:172402. [PMID: 31108054 DOI: 10.1016/j.ejphar.2019.05.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/07/2019] [Accepted: 05/16/2019] [Indexed: 12/18/2022]
Abstract
Anti-psychotic medications are widely used to treat schizophrenia and bipolar disorder. Besides their medical applications, anti-psychotic drugs have other pharmacological properties which are involved in multiple intracellular functions including metabolism, cell stress, cell-cycle regulation, survival and apoptosis through modulation of cellular signaling pathways such as PI3K/Akt/GSK-3β, STAT3 and wingless (Wnt)-related intracellular signaling. Also, anti-psychotics counteract the growth of tumor cells by stimulating the cellular immune system and natural killer cells. On the other hand, the positive charge and the lipophilicity of anti-psychotics have significant roles in the inhibition of P-gp pumps resulting in accumulation of chemotherapy drugs as well as increasing the cellular susceptibility to chemotherapy, autophagy, angiogenesis inhibition, stem cells differentiation induction and changing the expression of tumor suppressor genes and oncogenes. Overall, anti-psychotics are able to inhibit the proliferation of cancer cells through modulation of different cellular pathways. Anti-psychotics act as anti-cancer drugs and besides can increase the efficacy of anti-cancer agents in cancer cells. In this study, the anti-cancer effects of different anti-psychotic medicines on various malignant tumor cells and their molecular mechanisms have been discussed.
Collapse
|
12
|
Wang L, Pei J, Cong Z, Zou Y, Sun T, Davitt F, Garcia-Gil A, Holmes JD, O'Driscoll CM, Rahme K, Guo J. Development of anisamide-targeted PEGylated gold nanorods to deliver epirubicin for chemo-photothermal therapy in tumor-bearing mice. Int J Nanomedicine 2019; 14:1817-1833. [PMID: 30880982 PMCID: PMC6413758 DOI: 10.2147/ijn.s192520] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gold nanorods (AuNRs), due to the optical and electronic properties namely the surface plasma resonance, have been developed to achieve the light-mediated photothermal therapy (PTT) for cancer. However, PTT alone may suffer from inefficient tumor killing. Recently, the combination of PTT and chemotherapy has been utilized to achieve synergistic anticancer effects. METHODS In this study, AuNRs capped with hexadecyltrimethylammonium bromide (CTAB), poly(acrylic acid) (PAA), and PEGylated anisamide (a ligand known to target the sigma receptor) have been developed to produce a range of negatively charged anisamide-targeted PEGylated AuNRs (namely Au-CTAB-PAA-PEG-AA) for the combination of PTT and chemotherapy (termed as chemo-photothermal therapy [CPTT]). Epirubicin (EPI, an anthracycline drug) was efficiently loaded onto the surface of Au800-CTAB-PAA-PEG-AA via the electrostatic interaction forming Au800-CTAB-PAA-PEG-AA.EPI complex. RESULTS The resultant complex demonstrated pH-dependent drug release, facilitated nucleus trafficking of EPI, and induced antiproliferative effects in human prostate cancer PC-3 cells. When Au800-CTAB-PAA-PEG-AA.EPI complex was further stimulated with desired laser irradiation, the synergistic outcome was evident in PC-3 xenograft mice. CONCLUSION These results demonstrate a promising strategy for clinical application of CPTT in cancer.
Collapse
Affiliation(s)
- Limei Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China, ;
- Department of Pharmacy, The General Hospital of FAW, Changchun 130011, China
| | - Jin Pei
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China, ;
| | - Zhongcheng Cong
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China, ;
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China, ;
| | - Tianmeng Sun
- The First Hospital of Jilin University, Changchun 130021, China
| | - Fionán Davitt
- School of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland
- CRANN, Trinity College Dublin, Dublin, Ireland
| | - Adrià Garcia-Gil
- School of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland
- CRANN, Trinity College Dublin, Dublin, Ireland
| | - Justin D Holmes
- School of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland
- CRANN, Trinity College Dublin, Dublin, Ireland
| | | | - Kamil Rahme
- School of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland
- Department of Sciences, Faculty of Natural and Applied Science, Notre Dame University (Louaize), Zouk Mosbeh 1200, Lebanon
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China, ;
| |
Collapse
|
13
|
Luan X, Rahme K, Cong Z, Wang L, Zou Y, He Y, Yang H, Holmes JD, O'Driscoll CM, Guo J. Anisamide-targeted PEGylated gold nanoparticles designed to target prostate cancer mediate: Enhanced systemic exposure of siRNA, tumour growth suppression and a synergistic therapeutic response in combination with paclitaxel in mice. Eur J Pharm Biopharm 2019; 137:56-67. [PMID: 30779980 DOI: 10.1016/j.ejpb.2019.02.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/18/2018] [Accepted: 02/15/2019] [Indexed: 12/24/2022]
Abstract
Small interfering RNA (siRNA) has recently illustrated therapeutic potential for malignant disorders. However, the clinical application of siRNA-based therapeutics is significantly retarded by the paucity of successful delivery systems. Recently, multifunctional gold nanoparticles (AuNPs) as non-viral delivery carriers have shown promise for transporting chemotherapeutics, proteins/peptides, and genes. In this study, AuNPs capped with polyethylenimine (PEI) and PEGylated anisamide (a ligand known to target the sigma receptor) have been developed to produce a range of positively charged anisamide-targeted PEGylated AuNPs (namely Au-PEI-PEG-AA). The anisamide-targeted AuNPs effectively complexed siRNA via electrostatic interaction, and the resultant complex (Au110-PEI-PEG5000-AA.siRNA) illustrated favourable physicochemical characteristics, including particle size, surface charge, and stability. In vitro, anisamide-targeted AuNPs selectively bound to human prostate cancer PC-3 cells, inducing efficient endosomal escape of siRNA, and effective downregulation of the RelA gene. In vivo, prolonged systemic exposure of siRNA was achieved by anisamide-targeted AuNPs resulting in significant tumour growth suppression in a PC3 xenograft mouse model without an increase in toxicity. In addition, a combination of siRNA-mediated NF-κB knockdown using anisamide-targeted AuNPs with Paclitaxel produced a synergistic therapeutic response, thus providing a promising therapeutic strategy for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Xue Luan
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Kamil Rahme
- Department of Sciences, Faculty of Natural and Applied Science, Notre Dame University (Louaize), Zouk Mosbeh, Lebanon; Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland
| | - Zhongcheng Cong
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Limei Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Department of Pharmacy, The General Hospital of FAW, Changchun 130011, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yan He
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Hao Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Justin D Holmes
- Department of Chemistry and the Tyndall National Institute, University College Cork, Cork, Ireland; AMBER@CRANN, Trinity College Dublin, Dublin 2, Ireland
| | | | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
14
|
Tesei A, Cortesi M, Zamagni A, Arienti C, Pignatta S, Zanoni M, Paolillo M, Curti D, Rui M, Rossi D, Collina S. Sigma Receptors as Endoplasmic Reticulum Stress "Gatekeepers" and their Modulators as Emerging New Weapons in the Fight Against Cancer. Front Pharmacol 2018; 9:711. [PMID: 30042674 PMCID: PMC6048940 DOI: 10.3389/fphar.2018.00711] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
Despite the interest aroused by sigma receptors (SRs) in the area of oncology, their role in tumor biology remains enigmatic. The predominant subcellular localization and main site of activity of SRs are the endoplasmic reticulum (ER). Current literature data, including recent findings on the sigma 2 receptor subtype (S2R) identity, suggest that SRs may play a role as ER stress gatekeepers. Although SR endogenous ligands are still unknown, a wide series of structurally unrelated compounds able to bind SRs have been identified. Currently, the identification of novel antiproliferative molecules acting via SR interaction is a challenging task for both academia and industry, as shown by the fact that novel anticancer drugs targeting SRs are in the preclinical-stage pipeline of pharmaceutical companies (i.e., Anavex Corp. and Accuronix). So far, no clinically available anticancer drugs targeting SRs are still available. The present review focuses literature advancements and provides a state-of-the-art overview of SRs, with emphasis on their involvement in cancer biology and on the role of SR modulators as anticancer agents. Findings from preclinical studies on novel anticancer drugs targeting SRs are presented in brief.
Collapse
Affiliation(s)
- Anna Tesei
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Michela Cortesi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Alice Zamagni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Chiara Arienti
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Sara Pignatta
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRCCS), Meldola, Italy
| | - Mayra Paolillo
- Pharmacology Section, Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Daniela Curti
- Laboratory of Cellular and Molecular Neuropharmacology, Department of Biology and Biotechnology 'L. Spallanzani', University of Pavia, Pavia, Italy
| | - Marta Rui
- Medicinal Chemistry and Pharmaceutical Technology Section, Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Daniela Rossi
- Medicinal Chemistry and Pharmaceutical Technology Section, Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Simona Collina
- Medicinal Chemistry and Pharmaceutical Technology Section, Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
15
|
Xu YW, Zheng SB, Chen BS, Wen Y, Zhu SW. [Effect of sodium phenylbutyrate on the sensitivity of PC3/DTX-resistant prostate cancer cells to docetaxel]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 37:130-134. [PMID: 28109113 PMCID: PMC6765754 DOI: 10.3969/j.issn.1673-4254.2017.01.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To investigate the effect of sodium phenylbutyrate (SPB) in modulating docetaxel resistance in human prostate cancer cells in vitro. METHODS A PC3/docetaxel-resistant human prostate cancer cell line PC3/DTX was induced and examined for proliferation, viability, and cell inhibition rate in the presence of SPB. The concentration of concentration of docetaxel required to kill 50% of PC3/DTX cells incubated with 0, 1, 2, and 4 mmol/L SPB was determined using MTT assay. Cell apoptosis rate was analyzed with flow cytometry and the cellular expressions of p21, cyclin D1 and survivin proteins were detected using Western blotting. RESULTS Treatment of PC3/DTX cells with 0, 1, 2, and 4 mmol/L of SPB for 48 h resulted in cell viabilities of (99.85∓2.69)%, (84.68∓3.87)%, (68.65∓4.54)% and (43.54∓5.69)%, and cell inhibition rates of (10.69∓3.65)%, (25.78∓4.58)%, (54.68∓3.98)% and (69.84∓6.54)%, respectively (P<0.05). The concentration of docetaxel required to kill 50% of PC3/DTX cells cultured in the presence of with 0, 1, 2, and 4 mmol/L SPB was 135.98∓2.69, 109.65∓3.87, 87.65∓3.84 and 64.62∓2.98 nmol/L, respectively (P<0.05), and the cell apoptosis rates were (7.2∓0.8)%, (10.2∓0.9)%, (19.8∓2.1)% and (27.4∓2.5)%, respectively. SPB treatment promoted the protein expression of p21 and suppressed the expressions of cyclin D1 and survivin in PC3/DTX cells. CONCLUSION SPB can affect the expressions of p21, cyclin D1, and survivin in PC3/DTX cells and increase the sensitivity to the drug-resistant cells to docetaxel.
Collapse
Affiliation(s)
- Ya-Wen Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.E-mail:
| | | | | | | | | |
Collapse
|
16
|
Roney MSI, Park SK. Antipsychotic dopamine receptor antagonists, cancer, and cancer stem cells. Arch Pharm Res 2018; 41:384-408. [PMID: 29556831 DOI: 10.1007/s12272-018-1017-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 02/27/2018] [Indexed: 12/12/2022]
Abstract
Cancer is one of the deadliest diseases in the world. Despite extensive studies, treating metastatic cancers remains challenging. Years of research have linked a rare set of cells known as cancer stem cells (CSCs) to drug resistance, leading to the suggestion that eradication of CSCs might be an effective therapeutic strategy. However, few drug candidates are active against CSCs. New drug discovery is often a lengthy process. Drug screening has been advantageous in identifying drug candidates. Current understanding of cancer biology has revealed various clues to target cancer from different points of view. Many studies have found dopamine receptors (DRs) in various cancers. Therefore, DR antagonists have attracted a lot of attention in cancer research. Recently, a group of antipsychotic DR antagonists has been demonstrated to possess remarkable abilities to restrain and sensitize CSCs to existing chemotherapeutics by a process called differentiation approach. In this review, we will describe current aspects of CSC-targeting therapeutics, antipsychotic DR antagonists, and their extraordinary abilities to fight cancer.
Collapse
Affiliation(s)
- Md Saiful Islam Roney
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, Republic of Korea
| | - Song-Kyu Park
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, Republic of Korea.
- Research Driven Hospital, Korea University Guro Hospital, Biomedical Research Center, Seoul, 08308, Republic of Korea.
| |
Collapse
|
17
|
Arena E, Dichiara M, Floresta G, Parenti C, Marrazzo A, Pittalà V, Amata E, Prezzavento O. Novel Sigma-1 receptor antagonists: from opioids to small molecules: what is new? Future Med Chem 2018; 10:231-256. [PMID: 29185346 DOI: 10.4155/fmc-2017-0164] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023] Open
Abstract
Sigma-1 (σ1) receptor has been identified as a chaperone protein that interacts with other proteins, such as N-methyl-D-aspartate (NMDA) and opioid receptors, modulating their activity. σ1 receptor antagonists have been developed to obtain useful compounds for the treatment of psychoses, pain, drug abuse and cancer. Some interesting compounds such as E-5842 (5) and MS-377 (24), haloperidol and piperazine derivatives, respectively, were endowed with high affinity for σ1 receptors (Ki σ1 = 4 and 73 nM; Ki σ2 = 220 and 6900, respectively). They were developed for the treatment of psychotic disorders and 5 also underwent Phase II clinical trials suggesting interesting potential therapeutic applications. Here, σ1 receptor antagonists have been grouped based on chemical structure and reviewed according to structure-activity relationship and potential therapeutic role.
Collapse
Affiliation(s)
- Emanuela Arena
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Maria Dichiara
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Giuseppe Floresta
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
- Department of Chemical Sciences, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Carmela Parenti
- Department of Drug Sciences, Pharmacology Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Valeria Pittalà
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A Doria 6, 95125 Catania, Italy
| |
Collapse
|
18
|
Amata E, Rescifina A, Prezzavento O, Arena E, Dichiara M, Pittalà V, Montilla-García Á, Punzo F, Merino P, Cobos EJ, Marrazzo A. (+)-Methyl (1R,2S)-2-{[4-(4-Chlorophenyl)-4-hydroxypiperidin-1-yl]methyl}-1-phenylcyclopropanecarboxylate [(+)-MR200] Derivatives as Potent and Selective Sigma Receptor Ligands: Stereochemistry and Pharmacological Properties. J Med Chem 2017; 61:372-384. [PMID: 29220177 DOI: 10.1021/acs.jmedchem.7b01584] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Methoxycarbonyl-1-phenyl-2-cyclopropylmethyl based derivatives cis-(+)-1a [cis-(+)-MR200], cis-(-)-1a [cis-(-)-MR201], and trans-(±)-1a [trans-(±)-MR204], have been identified as new potent sigma (σ) receptor ligands. In the present paper, novel enantiomerically pure analogues were synthesized and optimized for their σ receptor affinity and selectivity. Docking studies rationalized the results obtained in the radioligand binding assay. Absolute stereochemistry was unequivocally established by X-ray analysis of precursor trans-(+)-5a as camphorsulfonyl derivative 9. The most promising compound, trans-(+)-1d, showed remarkable selectivity over a panel of more than 15 receptors as well as good chemical and enzymatic stability in human plasma. An in vivo evaluation evidenced that trans-(+)-1d, in contrast to trans-(-)-1d, cis-(+)-1d, or cis-(-)-1d, which behave as σ1 antagonists, exhibited a σ1 agonist profile. These data clearly demonstrated that compound trans-(+)-1d, due to its σ1 agonist activity and favorable receptor selectivity and stability, provided an useful tool for the study of σ1 receptors.
Collapse
Affiliation(s)
- Emanuele Amata
- Dipartimento di Scienze del Farmaco, Università di Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Antonio Rescifina
- Dipartimento di Scienze del Farmaco, Università di Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Dipartimento di Scienze del Farmaco, Università di Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuela Arena
- Dipartimento di Scienze del Farmaco, Università di Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Maria Dichiara
- Dipartimento di Scienze del Farmaco, Università di Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Valeria Pittalà
- Dipartimento di Scienze del Farmaco, Università di Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Ángeles Montilla-García
- Institute of Neuroscience and Department of Pharmacology, Faculty of Medicine, University of Granada , Avenida de Madrid 11, E-18012 Granada, Spain
| | - Francesco Punzo
- Dipartimento di Scienze del Farmaco, Università di Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Pedro Merino
- Laboratorio de Síntesis Asimétrica, Departamento de Síntesis y Estructura de Biomoléculas, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), Universidad de Zaragoza, CSIC , Campus San Francisco, E-50009 Zaragoza, Aragón, Spain
| | - Enrique J Cobos
- Institute of Neuroscience and Department of Pharmacology, Faculty of Medicine, University of Granada , Avenida de Madrid 11, E-18012 Granada, Spain
| | - Agostino Marrazzo
- Dipartimento di Scienze del Farmaco, Università di Catania , Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
19
|
Amata E, Dichiara M, Arena E, Pittalà V, Pistarà V, Cardile V, Graziano ACE, Fraix A, Marrazzo A, Sortino S, Prezzavento O. Novel Sigma Receptor Ligand-Nitric Oxide Photodonors: Molecular Hybrids for Double-Targeted Antiproliferative Effect. J Med Chem 2017; 60:9531-9544. [PMID: 29172528 DOI: 10.1021/acs.jmedchem.7b00791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This contribution reports the synthesis and evaluation of novel hybrid compounds that conjugate a sigma (σ) receptor pharmacophore and a nitric oxide (NO) photodonor. All compounds preserve their capability to generate NO under visible light and possess overall σ receptor nanomolar affinity, with one of them (8b) exhibiting remarkable σ2 receptor selectivity. Compounds 8b, 11a, and 11b were tested on tumorigenic MCF-7 and A2058 cells expressing high levels of σ2 and σ1 receptor, respectively. Considerable loss of cell viability was detected under light excitation, while negligible effects in the dark were detected. Moreover, they did not show any significant cytotoxicity in the dark or under irradiation on nontumorigenic NCTC-2544 keratinocytes. NO-induced reduction of cellular viability was demonstrated by in-cell NO detection and total nitrite estimation. For the first time, a combination of σ receptor moieties and a NO photodonor is reported, providing distinctive ligands potentially useful for cancer management.
Collapse
Affiliation(s)
- Emanuele Amata
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Maria Dichiara
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuela Arena
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Valeria Pittalà
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Venerando Pistarà
- Department of Drug Sciences, Laboratory of Organic Chemistry, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania , Via Santa Sofia 97, 95123 Catania, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania , Via Santa Sofia 97, 95123 Catania, Italy
| | - Aurore Fraix
- Department of Drug Sciences, Laboratory of Photochemistry, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Salvatore Sortino
- Department of Drug Sciences, Laboratory of Photochemistry, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
20
|
Longhitano L, Castracani CC, Tibullo D, Avola R, Viola M, Russo G, Prezzavento O, Marrazzo A, Amata E, Reibaldi M, Longo A, Russo A, Parrinello NL, Volti GL. Sigma-1 and Sigma-2 receptor ligands induce apoptosis and autophagy but have opposite effect on cell proliferation in uveal melanoma. Oncotarget 2017; 8:91099-91111. [PMID: 29207628 PMCID: PMC5710909 DOI: 10.18632/oncotarget.19556] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/13/2017] [Indexed: 01/30/2023] Open
Abstract
Uveal melanoma is the most common primary intraocular tumor in adults, with about 1200-1500 new cases occurring per year in the United States. Metastasis is a frequent occurrence in uveal melanoma, and outcomes are poor once distant spread occurs and no clinically significant chemotherapeutic protocol is so far available. The aim of the present study was to test the effect of various σ1 and σ2 receptor ligands as a possible pharmacological strategy for this rare tumor. Human uveal melanoma cells (92.1) were treated with various concentrations of different σ2 ligands (haloperidol and haloperidol metabolite II) and σ1 ligand ((+)-pentazocine) at various concentrations (1, 10 and 25 μM) and time points (0, 4 h, 8 h, 24 h and 48 h). Cell proliferation and migration were evaluated respectively by continuous cell monitoring by xCELLigence analysis, clonogenic assay and wound healing. Apoptosis and autophagy were also measured by cytofluorimetric and microscopy analysis. Our results showed that σ2 receptor ligands significantly reduced cell proliferation whereas (+)-pentazocine exhibited opposite results. All tested ligands showed significant decrease in cell migration. Interestingly, both σ1 and σ2 receptor ligands showed significant increase of autophagy and apoptosis at all concentrations. Taken all together these results suggest that sigma receptors mediates opposite biological effects but they also share common pharmacological effect on apoptosis and autophagy in uveal melanoma. In conclusion, these data provide the first evidence that sigma receptors may represent a "druggable" target to develop new chemotherapic agent for uveal melanoma.
Collapse
Affiliation(s)
- Lucia Longhitano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Roberto Avola
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Maria Viola
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuliano Russo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | | | - Emanuele Amata
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Michele Reibaldi
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Antonio Longo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Andrea Russo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Nunziatina Laura Parrinello
- Regional Reference Center for Rare Diseases, Clinical Division of Hematology and Transplantation, PO Ferrarotto Hospital, Azienda Ospedaliera-Universitaria Policlinico-Vittorio Emanuele, Via Citelli, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Euromediterranean Institute of Science and Technology, Palermo, Italy
| |
Collapse
|
21
|
Synthesis and evaluation of haloperidol metabolite II prodrugs as anticancer agents. Future Med Chem 2017; 9:1749-1764. [DOI: 10.4155/fmc-2017-0064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The use of haloperidol metabolite II (HP-metabolite II) prodrugs is an emerging strategy in the treatment of cancer. HP-metabolite II exhibits antiproliferative properties at micromolar concentrations inducing apoptosis in different types of cancer. Thus, the application of the prodrug approach appears as a useful method leading to much more desirable pharmacokinetic and pharmacodynamic properties. Some studies have shown that the esterification of the hydroxyl group of HP-metabolite II with 4-phenylbutiric acid (4-PBA) or valproic acid enhances the anticancer therapeutic potency. The current progresses in the design, synthesis and evaluation of anticancer activity of HP metabolite II prodrugs will be discussed in this review.
Collapse
|
22
|
Rescifina A, Floresta G, Marrazzo A, Parenti C, Prezzavento O, Nastasi G, Dichiara M, Amata E. Development of a Sigma-2 Receptor affinity filter through a Monte Carlo based QSAR analysis. Eur J Pharm Sci 2017; 106:94-101. [DOI: 10.1016/j.ejps.2017.05.061] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 10/19/2022]
|
23
|
Collina S, Bignardi E, Rui M, Rossi D, Gaggeri R, Zamagni A, Cortesi M, Tesei A. Are sigma modulators an effective opportunity for cancer treatment? A patent overview (1996-2016). Expert Opin Ther Pat 2017; 27:565-578. [PMID: 28051882 DOI: 10.1080/13543776.2017.1276569] [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] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Although several molecular targets against cancer have been identified, there is a continuous need for new therapeutic strategies. Sigma Receptors (SRs) overexpression has been recently associated with different cancer conditions. Therefore, novel anticancer agents targeting SRs may increase the specificity of therapies, overcoming some of the common drawbacks of conventional chemotherapy. Areas covered: The present review focuses on patent documents disclosing SR modulators with possible application in cancer therapy and diagnosis. The analysis reviews patents of the last two decades (1996-2016); patents were grouped according to target subtypes (S1R, S2R, pan-SRs) and relevant Applicants. The literature was searched through Espacenet, ISI Web, PatentScope and PubMed databases. Expert opinion: The number of patents related to SRs and cancer has increased in the last twenty years, confirming the importance of this receptor family as valuable target against neoplasias. Despite their short history in the cancer scenario, many SR modulators are at pre-clinical stage and one is undergoing a phase II clinical trial. SRs ligands may represent a powerful source of innovative antitumor therapeutics. Further investigation is needed for validating SR modulators as anti-cancer drugs. We strongly hope that this review could stimulate the interest of both Academia and pharmaceutical companies.
Collapse
Affiliation(s)
- Simona Collina
- a Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section , University of Pavia , Pavia , Italy
| | - Emanuele Bignardi
- a Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section , University of Pavia , Pavia , Italy
| | - Marta Rui
- a Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section , University of Pavia , Pavia , Italy
| | - Daniela Rossi
- a Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section , University of Pavia , Pavia , Italy
| | - Raffaella Gaggeri
- b Pharmacy Unit , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS , Meldola , Italy
| | - Alice Zamagni
- c Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS , Meldola , Italy
| | - Michela Cortesi
- c Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS , Meldola , Italy
| | - Anna Tesei
- d MBiochem, Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS , Meldola , Italy
| |
Collapse
|
24
|
Abstract
Sigma1 (also known as sigma-1 receptor, Sig1R, σ1 receptor) is a unique pharmacologically regulated integral membrane chaperone or scaffolding protein. The majority of publications on the subject have focused on the neuropharmacology of Sigma1. However, a number of publications have also suggested a role for Sigma1 in cancer. Although there is currently no clinically used anti-cancer drug that targets Sigma1, a growing body of evidence supports the potential of Sigma1 ligands as therapeutic agents to treat cancer. In preclinical models, compounds with affinity for Sigma1 have been reported to inhibit cancer cell proliferation and survival, cell adhesion and migration, tumor growth, to alleviate cancer-associated pain, and to have immunomodulatory properties. This review will highlight that although the literature supports a role for Sigma1 in cancer, several fundamental questions regarding drug mechanism of action and the physiological relevance of aberrant SIGMAR1 transcript and Sigma1 protein expression in certain cancers remain unanswered or only partially answered. However, emerging lines of evidence suggest that Sigma1 is a component of the cancer cell support machinery, that it facilitates protein interaction networks, that it allosterically modulates the activity of its associated proteins, and that Sigma1 is a selectively multifunctional drug target.
Collapse
Affiliation(s)
- Felix J Kim
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Philadelphia, PA, USA.
| | - Christina M Maher
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, USA
| |
Collapse
|
25
|
Olivieri M, Amata E, Vinciguerra S, Fiorito J, Giurdanella G, Drago F, Caporarello N, Prezzavento O, Arena E, Salerno L, Rescifina A, Lupo G, Anfuso CD, Marrazzo A. Antiangiogenic Effect of (±)-Haloperidol Metabolite II Valproate Ester [(±)-MRJF22] in Human Microvascular Retinal Endothelial Cells. J Med Chem 2016; 59:9960-9966. [PMID: 27739690 DOI: 10.1021/acs.jmedchem.6b01039] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
(±)-MRJF22 [(±)-2], a novel prodrug of haloperidol metabolite II (sigma-1 receptor antagonist/sigma-2 receptor agonist ligand) obtained by conjugation to valproic acid (histone deacetylase inhibitor) via an ester bond, exhibits antiangiogenic activity, being able to reduce human retinal endothelial cell (HREC) viability in a comparable manner to bevacizumab. Moreover, (±)-2 was able to significantly reduce viable cells count, endothelial cell migration, and tube formation in vascular endothelial growth factor A (VEGF-A) stimulated HREC cultures.
Collapse
Affiliation(s)
- Melania Olivieri
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Shila Vinciguerra
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Jole Fiorito
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Giovanni Giurdanella
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Nunzia Caporarello
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuela Arena
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Loredana Salerno
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Antonio Rescifina
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Carmelina Daniela Anfuso
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, University of Catania , Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
26
|
Zhou Y, Rucker EB, Zhou BP. Autophagy regulation in the development and treatment of breast cancer. Acta Biochim Biophys Sin (Shanghai) 2016; 48:60-74. [PMID: 26637829 DOI: 10.1093/abbs/gmv119] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/21/2015] [Indexed: 12/14/2022] Open
Abstract
Autophagy is a major catabolic process in which intracellular membrane structures, protein complexes, and lysosomes are formed as lysoautophagosome to degrade and renew cytoplasmic components. Autophagy is physiologically a strategy and mechanism for cellular homeostasis as well as adaptation to stress, and thus alterations in the autophagy machinery may lead to diverse pathological conditions. The role of autophagy in cancer is complex, and the current literature reflects this as a 'double-edged sword'. Autophagy shows promise as a novel therapeutic target in various types of breast cancer, inhibiting or increasing treatment efficacy in a context- and cell-type-dependent manner. This review aims to summarize the recent advances in the understanding of the mechanisms by which key modulators of autophagy participate in cancer metastasis, highlight different autophagy-deficient murine models for breast cancer study, and provide further impetus for the modulation of autophagy in anticancer therapy.
Collapse
Affiliation(s)
- Yuting Zhou
- Department of Molecular and Cellular Biochemistry, University of Kentucky School of Medicine, Lexington, KY 40506, USA Department of Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA
| | - Edmund B Rucker
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY 40506, USA Department of Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA
| | - Binhua P Zhou
- Department of Molecular and Cellular Biochemistry, University of Kentucky School of Medicine, Lexington, KY 40506, USA Department of Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY 40506, USA
| |
Collapse
|
27
|
van Waarde A, Rybczynska AA, Ramakrishnan NK, Ishiwata K, Elsinga PH, Dierckx RAJO. Potential applications for sigma receptor ligands in cancer diagnosis and therapy. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1848:2703-14. [PMID: 25173780 DOI: 10.1016/j.bbamem.2014.08.022] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/04/2014] [Accepted: 08/19/2014] [Indexed: 01/03/2023]
Abstract
Sigma receptors (sigma-1 and sigma-2) represent two independent classes of proteins. Their endogenous ligands may include the hallucinogen N,N-dimethyltryptamine (DMT) and sphingolipid-derived amines which interact with sigma-1 receptors, besides steroid hormones (e.g., progesterone) which bind to both sigma receptor subpopulations. The sigma-1 receptor is a ligand-regulated molecular chaperone with various ion channels and G-protein-coupled membrane receptors as clients. The sigma-2 receptor was identified as the progesterone receptor membrane component 1 (PGRMC1). Although sigma receptors are over-expressed in tumors and up-regulated in rapidly dividing normal tissue, their ligands induce significant cell death only in tumor tissue. Sigma ligands may therefore be used to selectively eradicate tumors. Multiple mechanisms appear to underlie cell killing after administration of sigma ligands, and the signaling pathways are dependent both on the type of ligand and the type of tumor cell. Recent evidence suggests that the sigma-2 receptor is a potential tumor and serum biomarker for human lung cancer and an important target for inhibiting tumor invasion and cancer progression. Current radiochemical efforts are focused on the development of subtype-selective radioligands for positron emission tomography (PET) imaging. Right now, the mostpromising tracers are [18F]fluspidine and [18F]FTC-146 for sigma-1 receptors and [11C]RHM-1 and [18F]ISO-1 for the sigma-2 subtype. Nanoparticles coupled to sigma ligands have shown considerable potential for targeted delivery of antitumor drugs in animal models of cancer, but clinical studies exploring this strategy in cancer patients have not yet been reported. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Anna A Rybczynska
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Nisha K Ramakrishnan
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Kiichi Ishiwata
- Tokyo Metropolitan Institute of Gerontology, Research Team for Neuroimaging, 35-2 Sakae-Cho, Itabashi-Ku, Tokyo 173-0015, Japan
| | - Philip H Elsinga
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; University of Ghent, University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
28
|
Sozio P, Fiorito J, Di Giacomo V, Di Stefano A, Marinelli L, Cacciatore I, Cataldi A, Pacella S, Turkez H, Parenti C, Rescifina A, Marrazzo A. Haloperidol metabolite II prodrug: asymmetric synthesis and biological evaluation on rat C6 glioma cells. Eur J Med Chem 2014; 90:1-9. [PMID: 25461306 DOI: 10.1016/j.ejmech.2014.11.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 11/04/2014] [Accepted: 11/05/2014] [Indexed: 01/27/2023]
Abstract
In a previous work we reported the antiproliferative effects of (±)-MRJF4, a novel haloperidol metabolite II (HP-mII) (a sigma-1 antagonist and sigma-2 agonist) prodrug, obtained through conjugation to 4-phenylbutyric acid (PhBA) [a histone deacetylase inhibitor (HDACi)] via an ester bond. As a continuation of this work, here we report the asymmetric synthesis of compounds (R)-(+)-MRJF4 and (S)-(-)-MRJF4 and the evaluation of their biological activity on rat C6 glioma cells, derived from glioblastoma multiforme (GBM), which is the most common and deadliest central nervous system (CNS) invasive malignancy. Favourable physicochemical properties, high permeability in the parallel artificial membrane permeability assay (PAMPA), good enzymatic and chemical stability, in vivo anticancer activity, associated with the capacity to reduce cell viability and to increase cell death by apoptosis, render compound (R)-(+)-MRJF4 a promising candidate for the development of a useful therapeutic for gliomas therapy.
Collapse
Affiliation(s)
- Piera Sozio
- Dipartimento di Farmacia, Università degli Studi di Chieti Gabriele D'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Jole Fiorito
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 630 W 168th St., New York, NY 10032, USA
| | - Viviana Di Giacomo
- Dipartimento di Farmacia, Università degli Studi di Chieti Gabriele D'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Antonio Di Stefano
- Dipartimento di Farmacia, Università degli Studi di Chieti Gabriele D'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Lisa Marinelli
- Dipartimento di Farmacia, Università degli Studi di Chieti Gabriele D'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Ivana Cacciatore
- Dipartimento di Farmacia, Università degli Studi di Chieti Gabriele D'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Amelia Cataldi
- Dipartimento di Farmacia, Università degli Studi di Chieti Gabriele D'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Stephanie Pacella
- Dipartimento di Farmacia, Università degli Studi di Chieti Gabriele D'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | - Hasan Turkez
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25240 Erzurum, Turkey
| | - Carmela Parenti
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Antonio Rescifina
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Dipartimento di Scienze del Farmaco, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy.
| |
Collapse
|
29
|
Kaushal N, Robson MJ, Rosen A, McCurdy CR, Matsumoto RR. Neuroprotective targets through which 6-acetyl-3-(4-(4-(4-fluorophenyl)piperazin-1-yl)butyl)benzo[d]oxazol-2(3H)-one (SN79), a sigma receptor ligand, mitigates the effects of methamphetamine in vitro. Eur J Pharmacol 2013; 724:193-203. [PMID: 24380829 DOI: 10.1016/j.ejphar.2013.12.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 11/16/2013] [Accepted: 12/12/2013] [Indexed: 10/25/2022]
Abstract
Exposure to high or repeated doses of methamphetamine can cause hyperthermia and neurotoxicity, which are thought to increase the risk of developing a variety of neurological conditions. Sigma receptor antagonism can prevent methamphetamine-induced hyperthermia and neurotoxicity, but the underlying cellular targets through which the neuroprotection is conveyed remain unknown. Differentiated NG108-15 cells were thus used as a model system to begin elucidating the neuroprotective mechanisms targeted by sigma receptor antagonists to mitigate the effects of methamphetamine. In differentiated NG108-15 cells, methamphetamine caused the generation of reactive oxygen/nitrogen species, an increase in PERK-mediated endoplasmic reticulum stress and the activation of caspase-3, -8 and -9, ultimately resulting in apoptosis at micromolar concentrations, and necrotic cell death at higher concentrations. The sigma receptor antagonist, 6-acetyl-3-(4-(4-(4-fluorophenyl)piperazin-1-yl)butyl)benzo[d]oxazol-2(3H)-one (SN79), attenuated methamphetamine-induced increases in reactive oxygen/nitrogen species, activation of caspase-3, -8 and -9 and accompanying cellular toxicity. In contrast, 1,3-di(2-tolyl)-guanidine (DTG), a sigma receptor agonist, shifted the dose response curve of methamphetamine-induced cell death towards the left. To probe the effect of temperature on neurotoxicity, NG108-15 cells maintained at an elevated temperature (40 °C) exhibited a significant and synergistic increase in cell death in response to methamphetamine, compared to cells maintained at a normal cell culture temperature (37 °C). SN79 attenuated the enhanced cell death observed in the methamphetamine-treated cells at 40 °C. Together, the data demonstrate that SN79 reduces methamphetamine-induced reactive oxygen/nitrogen species generation and caspase activation, thereby conveying neuroprotective effects against methamphetamine under regular and elevated temperature conditions.
Collapse
Affiliation(s)
- Nidhi Kaushal
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Matthew J Robson
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Abagail Rosen
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, School of Pharmacy, University of Mississippi, MS 38677, USA
| | - Rae R Matsumoto
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
30
|
Guo J, Ogier JR, Desgranges S, Darcy R, O′Driscoll C. Anisamide-targeted cyclodextrin nanoparticles for siRNA delivery to prostate tumours in mice. Biomaterials 2012; 33:7775-84. [DOI: 10.1016/j.biomaterials.2012.07.012] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 07/05/2012] [Indexed: 12/14/2022]
|
31
|
Kaushal N, Elliott M, Robson MJ, Iyer AKV, Rojanasakul Y, Coop A, Matsumoto RR. AC927, a σ receptor ligand, blocks methamphetamine-induced release of dopamine and generation of reactive oxygen species in NG108-15 cells. Mol Pharmacol 2012; 81:299-308. [PMID: 22101517 PMCID: PMC3286303 DOI: 10.1124/mol.111.074120] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 11/18/2011] [Indexed: 11/22/2022] Open
Abstract
Methamphetamine is a highly addictive psychostimulant drug of abuse that causes neurotoxicity with high or repeated dosing. Earlier studies demonstrated the ability of the selective σ receptor ligand N-phenethylpiperidine oxalate (AC927) to attenuate the neurotoxic effects of methamphetamine in vivo. However, the precise mechanisms through which AC927 conveys its protective effects remain to be determined. With the use of differentiated NG108-15 cells as a model system, the effects of methamphetamine on neurotoxic endpoints and mediators such as apoptosis, necrosis, generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS), and dopamine release were examined in the absence and presence of AC927. Methamphetamine at physiologically relevant micromolar concentrations caused apoptosis in NG108-15 cells. At higher concentrations of methamphetamine, necrotic cell death was observed. At earlier time points, methamphetamine caused ROS/RNS generation, which was detected with the fluorigenic substrate 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescin diacetate, acetyl ester, in a concentration- and time-dependent manner. N-Acetylcysteine, catalase, and l-N(G)-monomethyl arginine citrate inhibited the ROS/RNS fluorescence signal induced by methamphetamine, which suggests the formation of hydrogen peroxide and RNS. Exposure to methamphetamine also stimulated the release of dopamine from NG108-15 cells into the culture medium. AC927 attenuated methamphetamine-induced apoptosis, necrosis, ROS/RNS generation, and dopamine release in NG108-15 cells. Together, the data suggest that modulation of σ receptors can mitigate methamphetamine-induced cytotoxicity, ROS/RNS generation, and dopamine release in cultured cells.
Collapse
Affiliation(s)
- Nidhi Kaushal
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University Health Sciences Center, Morgantown, West Virginia 26506, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Marrazzo A, Cobos EJ, Parenti C, Aricò G, Marrazzo G, Ronsisvalle S, Pasquinucci L, Prezzavento O, Colabufo NA, Contino M, González LG, Scoto GM, Ronsisvalle G. Novel potent and selective σ ligands: evaluation of their agonist and antagonist properties. J Med Chem 2011; 54:3669-73. [PMID: 21476493 DOI: 10.1021/jm200144j] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Novel enantiomers and diastereoisomers structurally related to σ ligand (+)-MR200 were synthesized to improve σ(1)/σ(2) subtype selectivity. The selective σ(1) ligand (-)-8 showed an antagonist profile determined by phenytoin differential modulation of binding affinity in vitro, confirmed in vivo by an increase of κ opioid analgesia. The σ(2) ligand (-)-9 displayed agonist properties in an in vitro isolated organ bath assay and antiproliferative effects on LNCaP and PC3 prostate cancer cell lines.
Collapse
Affiliation(s)
- Agostino Marrazzo
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|