1
|
Zsoldos B, Nagy N, Donkó-Tóth V, Keglevich P, Weber M, Dékány M, Nehr-Majoros A, Szőke É, Helyes Z, Hazai L. Novel Piperazine Derivatives of Vindoline as Anticancer Agents. Int J Mol Sci 2024; 25:7929. [PMID: 39063170 PMCID: PMC11277489 DOI: 10.3390/ijms25147929] [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: 06/13/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
A series of novel vindoline-piperazine conjugates were synthesized by coupling 6 N-substituted piperazine pharmacophores at positions 10 and 17 of Vinca alkaloid monomer vindoline through different types of linkers. The in vitro antiproliferative activity of the 17 new conjugates was investigated on 60 human tumor cell lines (NCI60). Nine compounds presented significant antiproliferative effects. The most potent derivatives showed low micromolar growth inhibition (GI50) values against most of the cell lines. Among them, conjugates containing [4-(trifluoromethyl)benzyl]piperazine (23) and 1-bis(4-fluorophenyl)methyl piperazine (25) in position 17 of vindoline were outstanding. The first one was the most effective on the breast cancer MDA-MB-468 cell line (GI50 = 1.00 μM), while the second one was the most effective on the non-small cell lung cancer cell line HOP-92 (GI50 = 1.35 μM). The CellTiter-Glo Luminescent Cell Viability Assay was performed with conjugates 20, 23, and 25 on non-tumor Chinese hamster ovary (CHO) cells to determine the selectivity of the conjugates for cancer cells. These compounds exhibited promising selectivity with estimated half-maximal inhibitory concentration (IC50) values of 2.54 μM, 10.8 μM, and 6.64 μM, respectively. The obtained results may have an impact on the design of novel vindoline-based anticancer compounds.
Collapse
Affiliation(s)
- Bernadett Zsoldos
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Nóra Nagy
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Viktória Donkó-Tóth
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Péter Keglevich
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Márton Weber
- Spectroscopic Research Department, Gedeon Richter Plc., P.O. Box 27, H-1475 Budapest, Hungary
| | - Miklós Dékány
- Spectroscopic Research Department, Gedeon Richter Plc., P.O. Box 27, H-1475 Budapest, Hungary
| | - Andrea Nehr-Majoros
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary
- HUN-REN PTE Chronic Pain Research Group, H-7624 Pécs, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary
- HUN-REN PTE Chronic Pain Research Group, H-7624 Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- National Laboratory for Drug Research and Development, H-1117 Budapest, Hungary
- HUN-REN PTE Chronic Pain Research Group, H-7624 Pécs, Hungary
| | - László Hazai
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| |
Collapse
|
2
|
Latysheva AS, Zolottsev VA, Veselovsky AV, Scherbakov KA, Morozevich GE, Zhdanov DD, Novikov RA, Misharin AY. Oxazolinyl derivatives of androst-16-ene as inhibitors of CYP17A1 activity and prostate carcinoma cells proliferation: Effects of substituents in oxazolinyl moiety. J Steroid Biochem Mol Biol 2023; 230:106280. [PMID: 36870373 DOI: 10.1016/j.jsbmb.2023.106280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Steroid derivatives modified with nitrogen containing heterocycles are known to inhibit activity of steroidogenic enzymes, decrease proliferation of cancer cells and attract attention as promising anticancer agents. Specifically, 2'-(3β-hydroxyandrosta-5,16-dien-17-yl)-4',5'-dihydro-1',3'-oxazole 1a potently inhibited proliferation of prostate carcinoma cells. In this study we synthesized and investigated five new derivatives of 3β-hydroxyandrosta-5,16-diene comprising 4'-methyl or 4'-phenyl substituted oxazolinyl cycle 1 (b-f). Docking of compounds 1 (a-f) to CYP17A1 active site revealed that the presence of substitutents at C4' atom in oxazoline cycle, as well as C4' atom configuration, significantly affect docking poses of compounds in the complexes with enzyme. Testing of compounds 1 (a-f) as CYP17A1 inhibitors revealed that the only compound 1a, comprising unsubstituted oxazolinyl moiety, demonstrated strong inhibitory activity, while other compounds 1 (b-f) were slightly active or non active. Compounds 1 (a-f) efficiently decreased growth and proliferation of prostate carcinoma LNCaP and PC-3 cells at 96 h incubation; the effect of compound 1a was the most powerful. Compound 1a efficiently stimulated apoptosis and caused PC-3 cells death, that was demonstrated by a direct comparison of pro-apoptotic effects of compound 1a and abiraterone.
Collapse
Affiliation(s)
- Alexandra S Latysheva
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya street, 119435 Moscow, Russia
| | - Vladimir A Zolottsev
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya street, 119435 Moscow, Russia; RUDN University, 6, Miklukho-Maklaya street, 117198 Moscow, Russia.
| | - Alexander V Veselovsky
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya street, 119435 Moscow, Russia
| | - Kirill A Scherbakov
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya street, 119435 Moscow, Russia
| | - Galina E Morozevich
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya street, 119435 Moscow, Russia
| | - Dmitry D Zhdanov
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya street, 119435 Moscow, Russia; RUDN University, 6, Miklukho-Maklaya street, 117198 Moscow, Russia
| | - Roman A Novikov
- V.A. Engelhardt Institute of Molecular Biology RAS, 32, Vavilov street, Moscow, Russia
| | - Alexander Y Misharin
- V.N. Orekhovich Institute of Biomedical Chemistry, 10, Pogodinskaya street, 119435 Moscow, Russia
| |
Collapse
|
3
|
Design and Synthesis of New Agents for Prostate Cancer Treatment Inspired by Steroidal CYP17 A1 Inhibitors. ChemistrySelect 2022. [DOI: 10.1002/slct.202203393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
4
|
Khan II, Karshieva SS, Sokolova DV, Spirina TS, Zolottsev VA, Latysheva AS, Anisimova NY, Komarova MV, Yakunina MN, Nitetskaya TA, Misharin AY, Pokrovsky VS. Antiproliferative, proapoptotic, and tumor-suppressing effects of the novel anticancer agent alsevirone in prostate cancer cells and xenografts. Arch Pharm (Weinheim) 2021; 355:e2100316. [PMID: 34668210 DOI: 10.1002/ardp.202100316] [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: 08/26/2021] [Revised: 10/03/2021] [Accepted: 10/06/2021] [Indexed: 11/08/2022]
Abstract
The aim of this study was to explore the mechanisms of action of alsevirone in prostate cancer (PC) in vitro and in vivo: CYP17A1 inhibition, cytotoxic, apoptotic, and antitumor effects in comparison with abiraterone. The CYP17A1-inhibitory activity was investigated in rat testicular microsomes using high-performance liquid chromatography. Testosterone levels were evaluated using enzyme-linked immunoassay. IC50 values were calculated for PC3, DU-145, LNCaP, and 22Rv1 cells using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) test. The antitumor effect in vivo was studied in DU-145 and 22Rv1 subcutaneous xenografts in Balb/c nude mice. Alsevirone reduced the CYP17A1-inhibitory activity by 98% ± 0.2%. A statistically significant reduction in the testosterone concentration in murine blood was recorded after the 7th administration of 300 mg/kg alsevirone at 0.31 ± 0.03 ng/ml (p < .001) versus 0.98 ± 0.22 ng/ml (p = .392) after abiraterone administration and 1.52 ± 0.49 ng/ml in control animals. Alsevirone was more cytotoxic than abiraterone in DU-145, LNCaP, and 22Rv1 cells, with IC50 values of 23.80 ± 1.18 versus 151.43 ± 23.70 μM, 22.87 ± 0.54 versus 28.80 ± 1.61 μM, and 35.86 ± 5.63 versus 109.87 ± 35.15 μM, respectively. Alsevirone and abiraterone significantly increased annexin V-positive, caspase 3/7-positive, and activated Bcl-2-positive cells. In 22Rv1 xenografts, alsevirone 300 mg/kg × 10/24 h per os inhibited tumor growth: on Day 9 of treatment, tumor growth inhibition = 59% (p = .022). Thus, alsevirone demonstrated significant antitumor activity associated with CYP17A1 inhibition, apoptosis in PC cells, and testosterone reduction.
Collapse
Affiliation(s)
- Irina I Khan
- Laboratory of combined treatment, N. N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia.,Department of biochemistry, RUDN University, Moscow, Russia
| | - Saida S Karshieva
- Laboratory of combined treatment, N. N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia
| | - Darina V Sokolova
- Laboratory of combined treatment, N. N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia.,Department of biochemistry, RUDN University, Moscow, Russia
| | - Tatiana S Spirina
- Laboratory of combined treatment, N. N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia
| | - Vladimir A Zolottsev
- Department of biochemistry, RUDN University, Moscow, Russia.,Laboratory of synthesis of physiologically active compounds, Institute of Biomedical Chemistry, Moscow, Russia
| | - Alexandra S Latysheva
- Laboratory of synthesis of physiologically active compounds, Institute of Biomedical Chemistry, Moscow, Russia
| | - Natalia Y Anisimova
- Laboratory of combined treatment, N. N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia.,Laboratory of polymer materials, NUST "MISIS", Moscow, Russia
| | - Marina V Komarova
- Department of laser and biotechnical systems, Samara University, Samara, Russia
| | - Marina N Yakunina
- Laboratory of combined treatment, N. N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia
| | - Tatiana A Nitetskaya
- Laboratory of combined treatment, N. N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia
| | - Alexander Y Misharin
- Laboratory of synthesis of physiologically active compounds, Institute of Biomedical Chemistry, Moscow, Russia
| | - Vadim S Pokrovsky
- Laboratory of combined treatment, N. N. Blokhin National Medical Research Center of Oncology of Ministry of Health of Russian Federation, Moscow, Russia.,Department of biochemistry, RUDN University, Moscow, Russia.,Center of genetics and life sciences, Sirius University of Science and Technology, Sochi, Krasnodarsky Kray, Russia
| |
Collapse
|
5
|
Soylu H, Kırca M, Avcı S, Ozpolat B, Ustunel I. Antiandrogen abiraterone and docetaxel treatments affect Notch1, Jagged1 and Hes1 expressions in metastatic prostate cancer cells. Exp Mol Pathol 2021; 119:104607. [PMID: 33482170 DOI: 10.1016/j.yexmp.2021.104607] [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: 10/21/2020] [Revised: 12/10/2020] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Prostate cancer is the most common cancer in men. A Notch signaling pathway is an important pathway in cell proliferation, differentiation, and fate. However, currently, the effects of abiraterone based-anti-androgene therapy and docetaxel, the most commonly used standard chemotherapy in prostate cancer treatment, on Notch signaling pathway are unknown. This study aimed to investigate the effects of abiraterone acetate and docetaxel on the expression of Notch1, Jagged1 and Hes1 in prostate cancer cell lines. METHODS In vitro effects of abiraterone acetate and docetaxel were examined on Notch1, Jagged1, and Hes1 expression in LNCaP and PC3 PCa cell lines by immunofluorescence, Western blot, and qRT-PCR. MTT proliferation assay was used to evaluate cell proliferation and survival. RESULTS We found that in the treatment of PC3 cells with abiraterone acetate, docetaxel, and their combination, only mRNA expressions of Notch1, Jagged1 and Hes1 were affected compared to control, but these expression differences were not observed in protein expression. In LNCaP cells, abiraterone acetate and the combination groups reduced Notch1 protein expression. All treatment groups did not alter Jagged1 expression compared to control, but significantly increased the Hes1 gene and protein expression. CONCLUSION Our findings suggest that abiraterone and docetaxel treatments affects the expression of Notch signal pathway proteins. But these drugs especially cause significant upregulation in Hes1 expression in PCa cells. Therefore, co-application of Notch signaling inhibitors together with docetaxel and abiraterone chemotherapy, it was thought that decreased Hes1 expression could be stopped the deterioration of the prognosis of the patient.
Collapse
Affiliation(s)
- Hakan Soylu
- Department of Histology and Embryology, Faculty of Medicine, Duzce University, 81620 Duzce, Turkey
| | - Mustafa Kırca
- Department of Biochemistry, Faculty of Medicine, Kutahya Health Sciences University, 43100 Kutahya, Turkey
| | - Sema Avcı
- Department of Histology and Embryology, Faculty of Medicine, Alanya Alaaddin Keykubat University, 07070 Antalya, Turkey
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Ismail Ustunel
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, 07070 Antalya, TURKEY.
| |
Collapse
|
6
|
Kiss A, Herman BE, Görbe T, Mernyák E, Molnár B, Wölfling J, Szécsi M, Schneider G. Synthesis of novel 17-triazolyl-androst-5-en-3-ol epimers via Cu(I)-catalyzed azide-alkyne cycloaddition and their inhibitory effect on 17α-hydroxylase/C 17,20-lyase. Steroids 2018; 135:79-91. [PMID: 29577953 DOI: 10.1016/j.steroids.2018.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 03/15/2018] [Indexed: 11/18/2022]
Abstract
The regioselective Cu(I)-catalyzed 1,3-dipolar cycloaddition of 17α- and 17β-azidoandrost-5-en-3β-ol epimers (3b and 5b) with different terminal alkynes afforded novel 1,4-substituted triazolyl derivatives (8a-k and 9a-k). For the preparation of 5'-iodo-1',2',3'-triazoles (8m-n and 9m-n), an improved method was developed, directly from steroidal azides and terminal alkynes, in reaction mediated by CuI and ICl as iodinating agents. Acetolysis and subsequent hydrolysis of 8n and 9n yielded 5'-hydroxy-1',2',3'-triazoles 8o and 9o. The inhibitory effect of 8a-o, 9a-o, 3, and 5 on rat testicular C17,20-lyase was investigated by means of an in vitro radioincubation technique. The results revealed that the C-17 epimers of steroidal triazoles influence the C17,20-lyase effect. Inhibitors were found only in the 17α-triazolyl series (8a-o), whereas in the C-17 azide pair the 17β compound (5b) was more potent.
Collapse
Affiliation(s)
- Anita Kiss
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - Bianka Edina Herman
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10., H-6720 Szeged, Hungary
| | - Tamás Görbe
- Organic Chemistry, Arrhenius Laboratory, Stockholm University, 106 91 Stockholm, Sweden
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - Barnabás Molnár
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - János Wölfling
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary
| | - Mihály Szécsi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10., H-6720 Szeged, Hungary
| | - Gyula Schneider
- Department of Organic Chemistry, University of Szeged, Dóm tér 8., H-6720 Szeged, Hungary.
| |
Collapse
|
7
|
Li A, Yadav R, White JK, Herroon MK, Callahan BP, Podgorski I, Turro C, Scott EE, Kodanko JJ. Illuminating cytochrome P450 binding: Ru(ii)-caged inhibitors of CYP17A1. Chem Commun (Camb) 2018; 53:3673-3676. [PMID: 28304025 DOI: 10.1039/c7cc01459g] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
New Ru(ii)-caged abiraterone complexes were synthesized that exhibit strong absorption in the visible region and release the steroidal CYP17A1 inhibitor abiraterone upon exposure to low energy visible light in buffer and prostate cancer cells. Photoinduced release results in abiraterone binding to its CYP17A1 target in an inhibitory mode.
Collapse
Affiliation(s)
- Ao Li
- Department of Chemistry, Wayne State University, Detroit, MI 48202, USA.
| | - Rahul Yadav
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jessica K White
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, USA
| | - Mackenzie K Herroon
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan 48201, USA
| | - Brian P Callahan
- Department of Chemistry, Binghamton University, Binghamton, New York 13902, USA
| | - Izabela Podgorski
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan 48201, USA and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, USA
| | - Claudia Turro
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, USA
| | - Emily E Scott
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jeremy J Kodanko
- Department of Chemistry, Wayne State University, Detroit, MI 48202, USA. and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, USA
| |
Collapse
|
8
|
Li A, Turro C, Kodanko JJ. Ru(ii) polypyridyl complexes as photocages for bioactive compounds containing nitriles and aromatic heterocycles. Chem Commun (Camb) 2018; 54:1280-1290. [PMID: 29323683 PMCID: PMC5904840 DOI: 10.1039/c7cc09000e] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Photocaging allows for precise spatiotemporal control over the release of biologically active compounds with light. Most photocaged molecules employ organic photolabile protecting groups; however, biologically active compounds often contain functionalities such as nitriles and aromatic heterocycles that cannot be caged with organic groups. Despite their prevalence, only a few studies have reported successful caging of nitriles and aromatic heterocycles. Recently, Ru(ii)-based photocaging has emerged as a powerful method for the release of bioactive molecules containing these functional groups, in many cases providing high levels of spatial and temporal control over biological activity. This Feature Article discusses recent developments in applying Ru(ii)-based photocaging towards biological problems. Our groups designed and synthesized Ru(ii)-based platforms for the photoinduced delivery of cysteine protease and cytochrome P450 inhibitors in order to achieve selective control over enzyme inhibition. We also reported Ru(ii) photocaging groups derived from higher-denticity ancillary ligands that possess photophysical and photochemical properties distinct from more traditional Ru(ii)-based caging groups. In addition, for the first time, we are able to rapidly synthesize and screen Ru(ii) polypyridyl complexes that elicit desired properties by solid-phase synthesis. Finally, our work also defined steric and orbital mixing effects that are important factors in controlling photoinduced ligand exchange.
Collapse
Affiliation(s)
- Ao Li
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, Michigan 48202, USA.
| | | | | |
Collapse
|
9
|
Nesbitt H, Worthington J, Errington RJ, Patterson LH, Smith PJ, McKeown SR, McKenna DJ. The unidirectional hypoxia-activated prodrug OCT1002 inhibits growth and vascular development in castrate-resistant prostate tumors. Prostate 2017; 77:1539-1547. [PMID: 28944496 DOI: 10.1002/pros.23434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/09/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND OCT1002 is a unidirectional hypoxia-activated prodrug (uHAP) OCT1002 that can target hypoxic tumor cells. Hypoxia is a common feature in prostate tumors and is known to drive disease progression and metastasis. It is, therefore, a rational therapeutic strategy to directly target hypoxic tumor cells in an attempt to improve treatment for this disease. Here we tested OCT1002 alone and in combination with standard-of-care agents in hypoxic models of castrate-resistant prostate cancer (CRPC). METHODS The effect of OCT1002 on tumor growth and vasculature was measured using murine PC3 xenograft and dorsal skin fold (DSF) window chamber models. The effects of abiraterone, docetaxel, and cabazitaxel, both singly and in combination with OCT1002, were also compared. RESULTS The hypoxia-targeting ability of OCT1002 effectively controls PC3 tumor growth. The effect was evident for at least 42 days after exposure to a single dose (30 mg/kg) and was comparable to, or better than, drugs currently used in the clinic. In DSF experiments OCT1002 caused vascular collapse in the PC3 tumors and inhibited the revascularization seen in controls. In this model OCT1002 also enhanced the anti-tumor effects of abiraterone, cabazitaxel, and docetaxel; an effect which was accompanied by a more prolonged reduction in tumor vasculature density. CONCLUSIONS These studies provide the first evidence that OCT1002 can be an effective agent in treating hypoxic, castrate-resistant prostate tumors, either singly or in combination with established chemotherapeutics for prostate cancer.
Collapse
Affiliation(s)
- Heather Nesbitt
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| | | | - Rachel J Errington
- School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
- BioStatus Ltd, Shepshed, Leicestershire, United Kingdom
| | | | - Paul J Smith
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Stephanie R McKeown
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Declan J McKenna
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
10
|
Abstract
INTRODUCTION The hybridization of biologically active molecules is a powerful tool for drug discovery used to target a variety of diseases. It offers the prospect of better drugs for the treatment of a number of illnesses including cancer, malaria, tuberculosis and AIDS. Hybrid drugs can provide combination therapies in a single multi-functional agent and, by doing so, be more specific and powerful than conventional classic treatments. This research field is in great expansion and attracts many researchers worldwide. AREA COVERED This review covers the main research published between early 2013 to mid-2015 and takes into account several previous reviews on the subject. Its intention is to showcase the most recent advances reported towards the development of molecular hybrids in drug discovery. Particular attention is given to anticancer hybrids throughout the review. EXPERT OPINION Current advances show that molecular hybrids of biologically active molecules can lead to powerful therapeutics. Natural products play a key role in this field. It is also believed that toxin hybrids present a great opportunity for future progress and should be further explored. Furthermore, the synthesis of hybrid organometallics should be systematically studied as it can lead to potent drugs. The crucial requirement for growth still remains the efficacy of synthesis. Hence, the development of efficient synthetic methods allowing rapid access to diverse series of hybrids must be further investigated by researchers.
Collapse
Affiliation(s)
- Gervais Bérubé
- a Département de Chimie, Biochimie et Physique , Université du Québec à Trois-Rivières , Québec , Canada
| |
Collapse
|
11
|
Grossebrummel H, Peter T, Mandelkow R, Weiss M, Muzzio D, Zimmermann U, Walther R, Jensen F, Knabbe C, Zygmunt M, Burchardt M, Stope MB. Cytochrome P450 17A1 inhibitor abiraterone attenuates cellular growth of prostate cancer cells independently from androgen receptor signaling by modulation of oncogenic and apoptotic pathways. Int J Oncol 2015; 48:793-800. [PMID: 26648519 DOI: 10.3892/ijo.2015.3274] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/26/2015] [Indexed: 11/05/2022] Open
Abstract
Abiraterone provides significant survival advantages in prostate cancer (PC), however, the current understanding of the molecular mechanisms of abiraterone is still limited. Therefore, the abiraterone impact on androgen receptor (AR)-positive LNCaP and AR-negative PC-3 cells was assessed by cellular and molecular analyses. The present study demonstrated, that abiraterone treatment significantly decreased cell growth, AR expression, and AR activity of AR-positive LNCaP cells. Notably, AR-negative PC-3 cells exhibited comparable reductions in cellular proliferation, associated with DNA fragmentation and pro-apoptotic modulation of p21, caspase-3, survivin, and transforming growth factor β (TGFβ). Our observations suggest that the attenuation of AR signaling is not the only rationale to explain the abiraterone anticancer activity. Abiraterone efficacy may play a more global role in PC progression control than originally hypothesized. In this regard, abiraterone is not only a promising drug for treatment of AR-negative PC stages, even more, abiraterone may represent an alternative for treatment of other malignancies besides prostate cancer.
Collapse
Affiliation(s)
- Hannah Grossebrummel
- Department of Urology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Tilmann Peter
- Department of Urology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Robert Mandelkow
- Department of Urology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Martin Weiss
- Department of Urology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Damian Muzzio
- Department of Obstetrics and Gynaecology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Uwe Zimmermann
- Department of Urology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Reinhard Walther
- Department of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Federico Jensen
- Department of Obstetrics and Gynaecology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Cornelius Knabbe
- Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University Bochum, D-32545 Bad Oeynhausen, Germany
| | - Marek Zygmunt
- Department of Obstetrics and Gynaecology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Martin Burchardt
- Department of Urology, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, D-17475 Greifswald, Germany
| |
Collapse
|
12
|
Kovács D, Wölfling J, Szabó N, Szécsi M, Minorics R, Zupkó I, Frank É. Efficient access to novel androsteno-17-(1',3',4')-oxadiazoles and 17β-(1',3',4')-thiadiazoles via N-substituted hydrazone and N,N'-disubstituted hydrazine intermediates, and their pharmacological evaluation in vitro. Eur J Med Chem 2015; 98:13-29. [PMID: 25993309 DOI: 10.1016/j.ejmech.2015.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 04/13/2015] [Accepted: 05/06/2015] [Indexed: 01/05/2023]
Abstract
A series of novel 17-exo-oxadiazoles and -thiadiazoles in the Δ(5) androstene series were efficiently synthesized from pregnenolone acetate and pregnadienolone acetate via multistep pathways. 17β-(1',3',4')-Oxadiazoles were obtained in high yields by the phenyliodonium diacetate-induced oxidative ring closure of semicarbazone and N-acylhydrazones derived from 3β-acetoxy- and 3β-hydroxyandrost-5-ene-17β-carbaldehydes. For the synthesis of analogous Δ(16)-17-oxadiazolyl derivatives, N,N'-disubstituted hydrazine intermediates were prepared from 3β-acetoxyandrosta-5,16-diene-17-carboxylic acid, which then underwent cyclodehydration in the presence of POCl3. The cyclization of steroidal N,N'-diacylhydrazines containing a saturated ring D with the Lawesson reagent afforded 17β-(1',3',4')-thiadiazoles in good yields. Most of the products were subjected to deacetylation in basic media in order to enlarge the compound library available for pharmacological studies. All of these derivatives were screened in vitro for their antiproliferative effects against four malignant human adherent cell lines (HeLa, A2780, MCF7 and A431) by means of the MTT assay. The 3β-hydroxy derivatives of the newly-synthesized 17-exo-heterocycles were tested in vitro to investigate their inhibitory effects on rat testicular C17,20-lyase. One of the 1,3,4-oxadiazolyl derivatives proved to exert noteworthy enzyme-inhibitory action, with an IC50 (0.065 μM) of the same order of magnitude as that of abiraterone.
Collapse
Affiliation(s)
- Dóra Kovács
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - János Wölfling
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Nikoletta Szabó
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720 Szeged, Hungary
| | - Mihály Szécsi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720 Szeged, Hungary
| | - Renáta Minorics
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - István Zupkó
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Éva Frank
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| |
Collapse
|