1
|
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
|
2
|
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]
|
3
|
de la Calle CM, Shee K, Yang H, Lonergan PE, Nguyen HG. The endoplasmic reticulum stress response in prostate cancer. Nat Rev Urol 2022; 19:708-726. [PMID: 36168057 DOI: 10.1038/s41585-022-00649-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/09/2022]
Abstract
In order to proliferate in unfavourable conditions, cancer cells can take advantage of the naturally occurring endoplasmic reticulum-associated unfolded protein response (UPR) via three highly conserved signalling arms: IRE1α, PERK and ATF6. All three arms of the UPR have key roles in every step of tumour progression: from cancer initiation to tumour growth, invasion, metastasis and resistance to therapy. At present, no cure for metastatic prostate cancer exists, as targeting the androgen receptor eventually results in treatment resistance. New research has uncovered an important role for the UPR in prostate cancer tumorigenesis and crosstalk between the UPR and androgen receptor signalling pathways. With an improved understanding of the mechanisms by which cancer cells exploit the endoplasmic reticulum stress response, targetable points of vulnerability can be uncovered.
Collapse
Affiliation(s)
- Claire M de la Calle
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin Shee
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Heiko Yang
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Peter E Lonergan
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, St. James's Hospital, Dublin, Ireland
- Department of Surgery, Trinity College, Dublin, Ireland
| | - Hao G Nguyen
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Thomas E, Thankan RS, Purushottamachar P, Huang W, Kane MA, Zhang Y, Ambulos NP, Weber DJ, Njar VCO. Novel AR/AR-V7 and Mnk1/2 Degrader, VNPP433-3β: Molecular Mechanisms of Action and Efficacy in AR-Overexpressing Castration Resistant Prostate Cancer In Vitro and In Vivo Models. Cells 2022; 11:2699. [PMID: 36078112 PMCID: PMC9454942 DOI: 10.3390/cells11172699] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/20/2022] [Accepted: 08/27/2022] [Indexed: 11/29/2022] Open
Abstract
Prostate cancer (PCa) relies in part on AR-signaling for disease development and progression. Earlier, we developed drug candidate galeterone, which advanced through phase 2-clinical trials in treating castration-resistant PCa (CRPC). Subsequently, we designed, synthesized, and evaluated next-generation galeterone-analogs including VNPP433-3β which is potently efficacious against pre-clinical models of PCa. This study describes the mechanism of action of VNPP433-3β that promotes degradation of full-length AR (fAR) and its splice variant AR-V7 besides depleting MNK1/2 in in vitro and in vivo CRPC models that stably overexpresses fAR. VNPP433-3β directly engages AR within the cell and promotes proteasomal degradation of fAR and its splice variant AR-V7 by enhancing the interaction of AR with E3 ligases MDM2/CHIP but disrupting AR-HSP90 binding. Next, VNPP433-3β decreases phosphorylation of 4EBP1 and abates binding of eIF4E and eIF4G to 5' cap of mRNA by depleting MNK1/2 with consequent depletion of phosphorylated eIF4E. Finally, RNA-seq demonstrates modulation of multiple pathways that synergistically contribute to PCa inhibition. Therefore, VNPP433-3β exerts its antitumor effect by imposing 1) transcriptional regulation of AR and AR-responsive oncogenes 2) translational regulation by disrupting mRNA-5'cap-dependent translation initiation, 3) reducing AR half-life through enhanced proteasomal degradation in vitro and AR-overexpressing tumor xenografts in vivo.
Collapse
Affiliation(s)
- Elizabeth Thomas
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
- The Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
| | - Retheesh S. Thankan
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
- Flavocure Biotech, 701 E. Pratt St, Ste 2033, Baltimore, MD 21202, USA
- Isoprene Pharmaceuticals, Inc., 801 W Baltimore Street, Suite 502J, Baltimore, MD 21201, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
- The Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Yuji Zhang
- Division of Biostatistics and Bioinformatics, University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicholas P. Ambulos
- Department of Microbiology and Immunology, University of Maryland, Marlene Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| | - David J. Weber
- The Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Vincent C. O. Njar
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
- The Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA
| |
Collapse
|
5
|
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
|
6
|
Jin X, Yu R, Wang X, Proud CG, Jiang T. Progress in developing MNK inhibitors. Eur J Med Chem 2021; 219:113420. [PMID: 33892273 DOI: 10.1016/j.ejmech.2021.113420] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022]
Abstract
The MNKs (mitogen-activated protein kinase-interacting protein kinases) phosphorylate eIF4E (eukaryotic initiation factor 4 E) at serine 209; eIF4E plays an important role in the translation of cytoplasmic mRNAs, all of which possess a 5' 'cap' structure to which eIF4E binds. Elevated levels of eIF4E, p-eIF4E and/or the MNK protein kinases have been found in many types of cancer, including solid tumors and leukemia. MNKs also play a role in metabolic disease. Regulation of the activities of MNKs (MNK1 and MNK2), control the phosphorylation of eIF4E, which in turn has a close relationship with the processes of tumor development, cell migration and invasion, and energy metabolism. MNK knock-out mice display no adverse effects on normal cells or phenotypes suggesting that MNK may be a potentially safe targets for the treatment of various cancers. Several MNK inhibitors or 'degraders' have been identified. Initially, some of the inhibitors were developed from natural products or based on other protein kinase inhibitors which inhibit multiple kinases. Subsequently, more potent and selective inhibitors for MNK1/2 have been designed and synthesized. Currently, three inhibitors (BAY1143269, eFT508 and ETC-206) are in various stages of clinical trials for the treatment of solid cancers or leukemia, either alone or combined with inhibitors of other protein kinase. In this review, we summarize the diverse MNK inhibitors that have been reported in patents and other literature, including those with activities in vitro and/or in vivo.
Collapse
Affiliation(s)
- Xin Jin
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Rilei Yu
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xuemin Wang
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Christopher G Proud
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Tao Jiang
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
7
|
Latysheva AS, Zolottsev VA, Veselovsky AV, Scherbakov KA, Morozevich GE, Pokrovsky VS, Novikov RA, Timofeev VP, Tkachev YV, Misharin AY. New steroidal oxazolines, benzoxazoles and benzimidazoles related to abiraterone and galeterone. Steroids 2020; 153:108534. [PMID: 31678134 DOI: 10.1016/j.steroids.2019.108534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 10/01/2019] [Accepted: 10/25/2019] [Indexed: 02/06/2023]
Abstract
Seven new oxazoline, benzoxazole and benzimidazole derivatives were synthesized from 3β-acetoxyandrosta-5,16-dien-17-carboxylic, 3β-acetoxyandrost-5-en-17β-carboxylic and 3β-acetoxypregn-5-en-21-oic acids. Docking to active site of human 17α-hydroxylase/17,20-lyase revealed that all oxazolines, as well as benzoxazoles and benzimidazoles comprising Δ16 could form stable complexes with enzyme, in which steroid moiety is positioned similarly to that of abiraterone and galeterone, and nitrogen atom coordinates heme iron, while 16,17-saturated benzoxazoles and benzimidazoles could only bind in a position where heterocycle is located nearly parallel to heme plane. Modeling of the interaction of new benzoxazole and benzimidazole derivatives with androgen receptor revealed the destabilization of helix 12, constituting activation function 2 (AF2) site, by mentioned compounds, similar to one induced by known antagonist galeterone. The synthesized compounds inhibited growth of prostate carcinoma LNCaP and PC-3 cells at 96 h incubation; the potency of 2'-(3β-hydroxyandrosta-5,16-dien-17-yl)-4',5'-dihydro-1',3'-oxazole and 2'-(3β-hydroxyandrosta-5,16-dien-17-yl)-benzimidazole was superior and could inspire further investigations of these compounds as potential anti-cancer agents.
Collapse
Affiliation(s)
| | | | | | | | | | - Vadim S Pokrovsky
- Orekhovich Institute of Biomedical Chemistry, Moscow, Russia; N.N. Blokhin Cancer Research Center, Moscow, Russia; RUDN University, Moscow, Russia.
| | - Roman A Novikov
- Engelhardt Institute of Molecular Biology RAS, Moscow, Russia
| | | | | | | |
Collapse
|
8
|
In vitro antitumor activity, ADME-Tox and 3D-QSAR of synthesized and selected natural styryl lactones. Comput Biol Chem 2019; 83:107112. [DOI: 10.1016/j.compbiolchem.2019.107112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/13/2019] [Accepted: 08/18/2019] [Indexed: 12/13/2022]
|
9
|
Abazid A, Martin B, Choinowski A, McNeill RV, Brandenburg LO, Ziegler P, Zimmermann U, Burchardt M, Erb H, Stope MB. The androgen receptor antagonist enzalutamide induces apoptosis, dysregulates the heat shock protein system, and diminishes the androgen receptor and estrogen receptor β1 expression in prostate cancer cells. J Cell Biochem 2019; 120:16711-16722. [PMID: 31297844 DOI: 10.1002/jcb.28929] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 11/05/2022]
Abstract
Enzalutamide's accepted mode of action is by targeting the androgen receptor's (AR) activity. In clinical practice, enzalutamide demonstrates a good benefit-risk profile for the treatment of advanced prostate cancer (PC), even after poor response to standard antihormonal treatment. However, since both, well-established antiandrogens and enzalutamide, target AR functionality, we hypothesized that additional unknown mechanisms might be responsible for enzalutamide's superior anticancer activity. In the current study, PC cells were incubated with enzalutamide and enzalutamide-dependent modulation of apoptotic mechanisms were assessed via Western blot analysis, TDT-mediated dUTP-biotin nick end-labeling assay, and nuclear morphology assay. Alterations of heat shock protein (HSP), AR, and estrogen receptor (ER) expression were examined by Western blot analysis. Enzalutamide attenuated the proliferation of PC cells in a time- and dose-dependent manner. In the presence of enzalutamide, apoptosis occurred which was shown by increased BAX expression, decreased Bcl-2 expression, nuclear pyknosis, and genomic DNA fragmentation. Moreover, enzalutamide inhibited the expression of HSPs primarily involved in steroid receptor stabilization and suppressed AR and ERβ1 expression. This study demonstrates for the first time that enzalutamide treatment of PC cells triggers varying molecular mechanisms resulting in antiproliferative effects of the drug. In addition to the well-characterized antagonistic inhibition of AR functionality, we have shown that enzalutamide also affects the intracellular synthesis of steroid receptor-associated HSPs, thereby diminishing the expression of AR and ERβ1 proteins and inducing apoptotic pathways. According to an indirect attenuation of HSP-associated factors such as steroid receptors, endometrial carcinoma, uterine leiomyosarcoma, and mamma carcinoma cells also demonstrated inhibited cell growth in the presence of enzalutamide. Our data, therefore, suggest that enzalutamide's high efficacy is at least partially independent of AR and p53 protein expression, which are frequently lost in advanced PC.
Collapse
Affiliation(s)
- Alexander Abazid
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Benedikt Martin
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Anja Choinowski
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Rhiannon V McNeill
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Frankfurt, Frankfurt, Germany
| | | | - Patrick Ziegler
- Institute for Occupational and Social Medicine, RWTH Aachen University, Aachen, Germany
| | - Uwe Zimmermann
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Martin Burchardt
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | - Holger Erb
- Department of Urology, University of Dresden, Dresden, Germany
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
10
|
Cai X, Zhao S, Cai D, Zheng J, Zhu Z, Wei D, Zheng Z, Zhu H, Chen Y. Synthesis and evaluation of novel D-ring substituted steroidal pyrazolines as potential anti-inflammatory agents. Steroids 2019; 146:70-78. [PMID: 30951758 DOI: 10.1016/j.steroids.2019.03.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/06/2019] [Accepted: 03/27/2019] [Indexed: 02/05/2023]
Abstract
To identify new potential anti-inflammatory agents, a number of novel steroidal derivatives with nitrogen heterocyclic side chains 4a-4l were synthesized and evaluated for their anti-inflammatory effects in activated RAW 264.7 macrophage cells. The synthesis scheme involves two steps, Claisen-Schmidt condensation with the corresponding pregnenolone and aromatic aldehydes as the first step followed by nucleophilic addition of thiosemicarbazide across an α, β-unsaturated carbonyl as a later step. Compound structures were confirmed by 1H NMR, 13C NMR, HRMS, and IR. The compounds were assayed to test their anti-inflammatory effects in activated RAW 264.7 cells. Compound 4g, 3β-hydroxy-pregn-5-en-17β-yl-5'-(m-fluorophenyl)-4', 5'-dihydro-1'-carbothioic acid amido pyrazole, was identified as the most potent anti-inflammatory agent of the analysed compounds, with an IC50 value of 0.86 µM on nitric oxide (NO) production in lipopolysaccharide (LPS)-induced RAW 264.7 cells for 24 h compared to dexamethasone (IC50 = 0.62 µM) and low cytotoxicity against RAW 264.7 cells. Compound 4g significantly inhibited NO produced by LPS-induced RAW 264.7 cells. Further studies showed that compound 4g markedly inhibited the expression of pro-inflammatory factors, including inducible nitric oxide synthase (iNOS), interleukin-6 (IL-6), tumour necrosis factor-α (TNF-α), and cyclooxygenase-2 (COX-2) in LPS-induced RAW 264.7 cells. These results indicate that derivatives bearing pyrazoline structure might be considered for further research and scaffold optimization in designing anti-inflammatory drugs and compound 4g might be a promising therapeutic anti-inflammatory drug candidate.
Collapse
Affiliation(s)
- Xiaorui Cai
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Shulin Zhao
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - De Cai
- Department of Pharmacy, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jinhong Zheng
- Department of Chemistry, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Zhiwei Zhu
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Duncan Wei
- Department of Pharmacy, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Zhiwei Zheng
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Huide Zhu
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yicun Chen
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China.
| |
Collapse
|
11
|
Brito V, Santos AO, Almeida P, Silvestre S. Novel 4-azaandrostenes as prostate cancer cell growth inhibitors: Synthesis, antiproliferative effects, and molecular docking studies. CR CHIM 2019. [DOI: 10.1016/j.crci.2018.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
12
|
Dong J, Wu Z, Wang D, Pascal LE, Nelson JB, Wipf P, Wang Z. Hsp70 Binds to the Androgen Receptor N-terminal Domain and Modulates the Receptor Function in Prostate Cancer Cells. Mol Cancer Ther 2018; 18:39-50. [PMID: 30297360 DOI: 10.1158/1535-7163.mct-18-0432] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/15/2018] [Accepted: 10/03/2018] [Indexed: 01/03/2023]
Abstract
The androgen receptor (AR) is a key driver and therapeutic target in androgen-sensitive prostate cancer, castration-resistant prostate cancer (CRPC), and CRPC resistant to abiraterone and enzalutamide, two second-generation inhibitors of AR signaling. Because current AR inhibitors target a functioning C-terminal ligand-binding domain (LBD), the identification and characterization of cofactors interacting with the N-terminal domain (NTD) of AR may lead to new approaches to target AR signaling in CRPC. Using a pull-down approach coupled with proteomics, we have identified Hsp70 as a cofactor for the NTD of AR in prostate cancer cells. Hsp70 inhibition using siRNA or small molecules indicated that Hsp70 played an important role in the expression and transactivation of endogenous AR. Prostate-specific antigen (PSA) promoter/enhancer-driven luciferase assays showed that Hsp70 was also required for transactivation of AR mutant lacking LBD. Furthermore, clonogenic assays showed that an Hsp70 inhibitor, either alone or in synergy with enzalutamide, can inhibit the proliferation of 22Rv1, a widely used enzalutamide-resistant CRPC prostate cancer cell line. These findings suggest that Hsp70 is a potential therapeutic target for the treatment of enzalutamide-resistant CRPC.
Collapse
Affiliation(s)
- Jun Dong
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeyu Wu
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dan Wang
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Laura E Pascal
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Joel B Nelson
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Peter Wipf
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhou Wang
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. .,University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
13
|
Oxazoline derivatives of [17(20)E]-21-norpregnene – inhibitors of CYP17A1 activity and proliferation of prostate carcinoma cells. Russ Chem Bull 2018. [DOI: 10.1007/s11172-018-2122-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
14
|
Savić MP, Ajduković JJ, Plavša JJ, Bekić SS, Ćelić AS, Klisurić OR, Jakimov DS, Petri ET, Djurendić EA. Evaluation of A-ring fused pyridine d-modified androstane derivatives for antiproliferative and aldo-keto reductase 1C3 inhibitory activity. MEDCHEMCOMM 2018; 9:969-981. [PMID: 30108986 DOI: 10.1039/c8md00077h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/27/2018] [Indexed: 01/22/2023]
Abstract
New A-ring pyridine fused androstanes in 17a-homo-17-oxa (d-homo lactone), 17α-picolyl or 17(E)-picolinylidene series were synthesized and validated by X-ray crystallography, HRMS, IR and NMR spectroscopy. Novel compounds 3, 5, 8 and 12 were prepared by treatment of 4-en-3-one or 4-ene-3,6-dione d-modified androstane derivatives with propargylamine catalyzed by Cu(ii), and evaluated for potential anticancer activity in vitro using human cancer cell lines and recombinant targets of steroidal anti-cancer drugs. Pyridine fusion to position 3,4 of the A-ring may dramatically enhance affinity of 17α-picolyl compounds for CYP17 while conferring selective antiproliferative activity against PC-3 cells. Similarly, pyridine fusion to the A-ring of steroidal d-homo lactones led to identification of new inhibitors of aldo-keto reductase 1C3, an enzyme targeted in acute myeloid leukemia, breast and prostate cancers. One A-pyridine d-lactone steroid 5 also has selective submicromolar antiproliferative activity against HT-29 colon cancer cells. None of the new derivatives have affinity for estrogen or androgen receptors in a yeast screen, suggesting negligible estrogenicity and androgenicity. Combined, our results suggest that A-ring pyridine fusions have potential in modulating the anticancer activity of steroidal compounds.
Collapse
Affiliation(s)
- Marina P Savić
- Department of Chemistry, Biochemistry and Environmental Protection , Faculty of Sciences , University of Novi Sad , Trg Dositeja Obradovića 3 , 21000 Novi Sad , Serbia .
| | - Jovana J Ajduković
- Department of Chemistry, Biochemistry and Environmental Protection , Faculty of Sciences , University of Novi Sad , Trg Dositeja Obradovića 3 , 21000 Novi Sad , Serbia .
| | - Jovana J Plavša
- Department of Biology and Ecology , Faculty of Sciences , University of Novi Sad , Trg Dositeja Obradovića 2 , 21000 Novi Sad , Serbia .
| | - Sofija S Bekić
- Department of Chemistry, Biochemistry and Environmental Protection , Faculty of Sciences , University of Novi Sad , Trg Dositeja Obradovića 3 , 21000 Novi Sad , Serbia .
| | - Andjelka S Ćelić
- Department of Biology and Ecology , Faculty of Sciences , University of Novi Sad , Trg Dositeja Obradovića 2 , 21000 Novi Sad , Serbia .
| | - Olivera R Klisurić
- Department of Physics , Faculty of Sciences , University of Novi Sad , Trg Dositeja Obradovića 4 , 21000 Novi Sad , Serbia
| | - Dimitar S Jakimov
- Oncology Institute of Vojvodina , Faculty of Medicine , University of Novi Sad , Put Dr Goldmana 4 , 21204 Sremska Kamenica , Serbia
| | - Edward T Petri
- Department of Biology and Ecology , Faculty of Sciences , University of Novi Sad , Trg Dositeja Obradovića 2 , 21000 Novi Sad , Serbia .
| | - Evgenija A Djurendić
- Department of Chemistry, Biochemistry and Environmental Protection , Faculty of Sciences , University of Novi Sad , Trg Dositeja Obradovića 3 , 21000 Novi Sad , Serbia .
| |
Collapse
|
15
|
Abiraterone and Ionizing Radiation Alter the Sphingolipid Homeostasis in Prostate Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1112:293-307. [DOI: 10.1007/978-981-13-3065-0_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
16
|
Zolottsev VA, Tkachev YV, Latysheva AS, Kostin VA, Novikov RA, Timofeev VP, Morozevich GE, Kuzikov AV, Shumyantseva VV, Misharin AY. Comparison of [17(20)E]-21-Norpregnene oxazolinyl and benzoxazolyl derivatives as inhibitors of CYP17A1 activity and prostate carcinoma cells growth. Steroids 2018; 129:24-34. [PMID: 29183745 DOI: 10.1016/j.steroids.2017.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/09/2017] [Accepted: 11/15/2017] [Indexed: 01/02/2023]
Abstract
Four new 4,5-dihydro-1,3-oxazole, and four new benzo-[d]-oxazole derivatives of [17(20)E]-21-norpregnene, differing in the structure of steroid moiety, were synthesized and evaluated for their potency to inhibit 17α-hydroxylase/17,20-lyase (CYP17A1) activity. Among new compounds, the only oxazolinyl derivative comprising 5-oxo-4,5-seco-3-yn- moiety potently inhibited CYP17A1. Binding modes of the oxazolinyl derivatives of [17(20)E]-21-norpregnene were analyzed by molecular dynamics simulations, and model of alternate, water-bridged type II interaction was proposed for these compounds. Eight new compounds, together with two CYP17A1-inhibiting oxazolinyl derivatives synthesized earlier, abiraterone and galeterone were evaluated for their potency to inhibit prostate carcinoma PC-3 and LNCaP cells growth. Oxazolinyl and benzoxazolyl derivatives comprising 3β-hydroxy-5-ene moieties potently inhibited prostate carcinoma cell growth; inhibitory potencies of 3-oxo-4-en- and 5-oxo-4,5-seco-3-yn- derivatives were significantly lower.
Collapse
Affiliation(s)
| | | | | | | | - Roman A Novikov
- Engelhardt Institute of Molecular Biology RAS, Moscow, Russia
| | | | | | | | | | | |
Collapse
|
17
|
Romero-Hernández LL, Merino-Montiel P, Meza-Reyes S, Vega-Baez JL, López Ó, Padrón JM, Montiel-Smith S. Synthesis of unprecedented steroidal spiro heterocycles as potential antiproliferative drugs. Eur J Med Chem 2017; 143:21-32. [PMID: 29172080 DOI: 10.1016/j.ejmech.2017.10.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/09/2017] [Accepted: 10/22/2017] [Indexed: 01/14/2023]
Abstract
Herein we report the straightforward preparation of novel conformationally-restricted steroids from trans-androsterone and estrone, decorated with spiranic oxazolidin-2-one or 2-aminooxazoline motifs at C-17 as potential antiproliferative agents. Such unprecedented pharmacophores were accessed using an aminomethylalcohol derivative at C-17 as the key intermediate; reaction of such functionality with triphosgene, or conversion into N-substituted thioureas, followed by an intramolecular cyclodesulfurization reaction promoted by yellow HgO, furnished such spirocycles in excellent yields. Title compounds were tested in vitro against a panel of six human tumor cell lines, named A549 (non-small cell lung), HBL-100 (breast), HeLa (cervix), SW1573 (non-small cell lung), T-47D (breast) and WiDr (colon), and the results were compared with steroidal chemotherapeutic agents (abiraterone and galeterone); the A-ring of the steroidal backbone, the nature of the heterocycle and the N-substituents proved to be essential motifs for establishing structure-activity relationships concerning not only the potency but also the selectivity against tumor cell lines. Estrone derivatives, particularly those bearing a spiranic 2-aminooxazoline scaffold were found to be the most active compounds, with GI50 values ranging from the low micromolar to the submicromolar level (0.34-1.5 μM). Noteworthy, the lead compounds showed a remarkable increase in activity against the resistant cancer cell lines (T-47D and WiDr) compared to the anticancer reference drugs (up to 120-fold).
Collapse
Affiliation(s)
- Laura L Romero-Hernández
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico
| | - Penélope Merino-Montiel
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico.
| | - Socorro Meza-Reyes
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico
| | - José Luis Vega-Baez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, E-41071 Seville, Spain
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Centro de Investigaciones Biomédicas de Canarias (CIBICAN), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - Sara Montiel-Smith
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Pue., Mexico.
| |
Collapse
|
18
|
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
|
19
|
McCarty DJ, Huang W, Kane MA, Purushottamachar P, Gediya LK, Njar VC. Novel galeterone analogs act independently of AR and AR-V7 for the activation of the unfolded protein response and induction of apoptosis in the CWR22Rv1 prostate cancer cell model. Oncotarget 2017; 8:88501-88516. [PMID: 29179452 PMCID: PMC5687622 DOI: 10.18632/oncotarget.19762] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/06/2017] [Indexed: 12/29/2022] Open
Abstract
The androgen receptor (AR) has long been the primary target for the treatment of prostate cancer (PC). Despite continuous efforts to block AR activity through ligand depletion, AR antagonism, AR depletion and combinations thereof, advanced PC tumors remain resilient. Herein, we evaluate two galeterone analogs, VNPT-178 and VNLG-74A, in PC cell models of diverse androgen and AR dependence attempting to delineate their mechanisms of action and potential clinical utility. Employing basic biochemical techniques, we determined that both analogs have improved antiproliferative and anti-AR activities compared to FDA-approved abiraterone and enzalutamide. However, induction of apoptosis in these models is independent of the AR and its truncated variant, AR-V7, and instead likely results from sustained endoplasmic reticulum stress and deregulated calcium homeostasis. Using in silico molecular docking, we predict VNPT-178 and VNLG-74A bind the ATPase domain of BiP/Grp78 and Hsp70-1A with greater affinity than the AR. Disruption of 70 kDa heat shock protein function may be the underlying mechanism of action for these galeterone analogs. Therefore, despite simultaneously antagonizing AR activity, AR and/or AR-V7 expression alone may inadequately predict a patient's response to treatment with VNPT-178 or VNLG-74A. Future studies evaluating the context-specific limitations of these compounds may provide clarity for their clinical application.
Collapse
Affiliation(s)
- David J. McCarty
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lalji K. Gediya
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Vincent C.O. Njar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
20
|
Njar VCO. Androgen receptor antagonism and impact on inhibitors of androgen synthesis in prostate cancer therapy. Transl Cancer Res 2017; 6:S1128-S1131. [PMID: 30613487 DOI: 10.21037/tcr.2017.08.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA.,Marlene Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Kwegyir-Afful AK, Murigi FN, Purushottamachar P, Ramamurthy VP, Martin MS, Njar VCO. Galeterone and its analogs inhibit Mnk-eIF4E axis, synergize with gemcitabine, impede pancreatic cancer cell migration, invasion and proliferation and inhibit tumor growth in mice. Oncotarget 2016; 8:52381-52402. [PMID: 28881737 PMCID: PMC5581036 DOI: 10.18632/oncotarget.14154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/19/2016] [Indexed: 12/22/2022] Open
Abstract
Survival rate for pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) is poor, with about 80% of patients presenting with the metastatic disease. Gemcitabine, the standard chemotherapeutic agent for locally advanced and metastatic PDAC has limited efficacy, attributed to innate/acquired resistance and activation of pro-survival pathways. The Mnk1/2-eIF4E and NF-κB signaling pathways are implicated in PDAC disease progression/metastasis and also associated with gemcitabine-induced resistance in PDAC. Galeterone (gal), a multi-target, agent in phase III clinical development for prostate cancer has also shown effects on the aforementioned pathways. We show for the first time, that gal/analogs (VNPT55, VNPP414 and VNPP433-3β) profoundly inhibited cell viability of gemcitabine-naive/resistance PDAC cell lines and strongly synergized with gemcitabine in gemcitabine-resistant PDAC cells. In addition, to inducing G1 cell cycle arrest, gal/analogs induced caspase 3-mediated cell-death of PDAC cells. Gal/analogs caused profound downregulation of Mnk1/2, peIF4E and NF-κB (p-p65), metastatic inducing factors (N-cadherin, MMP-1/-2/-9, Slug, Snail and CXCR4) and putative stem cell factors, (β-Catenin, Nanog, BMI-1 and Oct-4). Gal/analog also depleted EZH2 and upregulated E-Cadherin. These effects resulted in significant inhibition of PDAC cell migration, invasion and proliferation. Importantly, we also observed strong MiaPaca-2 tumor xenograft growth inhibition (61% to 92%). Collectively, these promising findings strongly support further development of gal/analogs as novel therapeutics for PDAC.
Collapse
Affiliation(s)
- Andrew K Kwegyir-Afful
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Francis N Murigi
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Vidya P Ramamurthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Marlena S Martin
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Current Address: Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, VA 20147, USA
| | - Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| |
Collapse
|
22
|
Kovačević SZ, Podunavac-Kuzmanović SO, Jevrić LR, Vukić VR, Savić MP, Djurendić EA. Preselection of A- and B- modified d-homo lactone and d-seco androstane derivatives as potent compounds with antiproliferative activity against breast and prostate cancer cells - QSAR approach and molecular docking analysis. Eur J Pharm Sci 2016; 93:107-13. [PMID: 27503457 DOI: 10.1016/j.ejps.2016.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/29/2016] [Accepted: 08/04/2016] [Indexed: 11/19/2022]
Abstract
The problem with trial-and-error approach in organic synthesis of targeted anticancer compounds can be successfully avoided by computational modeling of molecules, docking studies and chemometric tools. It has been proven that A- and B- modified d-homo lactone and d-seco androstane derivatives are compounds with significant antiproliferative activity against estrogen-independent breast adenocarcinoma (ER-, MDA-MB-231) and androgen-independent prostate cancer cells (AR-, PC-3). This paper presents the quantitative structure-activity relationship (QSAR) models based on artificial neural networks (ANNs) which are able to predict whether d-homo lactone and/or d-seco androstane-based compounds will express antiproliferative activity against breast cancer cells (MDA-MB-231) or not. Also, the present paper describes the molecular docking study of 3β-acetoxy-5α,6α-epoxy- (3) and 6α,7α-epoxy-1,4-dien-3-one (24) d-homo lactone androstane derivatives, as well as 4-en-3-one (15) d-seco androstane derivative, which are compounds with strong or moderate antiproliferative activity against prostate cancer cells (PC-3), and compares them with commercially available medicament for prostate cancer - abiraterone. The obtained promising results can be used as guidelines in further syntheses of novel d-homo lactone and d-seco androstane derivatives with antiproliferative activity against breast and prostate cancer cells.
Collapse
Affiliation(s)
- Strahinja Z Kovačević
- University of Novi Sad, Faculty of Technology Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia.
| | | | - Lidija R Jevrić
- University of Novi Sad, Faculty of Technology Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia
| | - Vladimir R Vukić
- University of Novi Sad, Faculty of Technology Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia
| | - Marina P Savić
- University of Novi Sad, Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| | - Evgenija A Djurendić
- University of Novi Sad, Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| |
Collapse
|
23
|
Kwegyir-Afful AK, Bruno RD, Purushottamachar P, Murigi FN, Njar VCO. Galeterone and VNPT55 disrupt Mnk-eIF4E to inhibit prostate cancer cell migration and invasion. FEBS J 2016; 283:3898-3918. [PMID: 27618366 DOI: 10.1111/febs.13895] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/16/2016] [Accepted: 09/09/2016] [Indexed: 12/28/2022]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) accounts for a high percentage of prostate cancer mortality. The proprietary compound galeterone (gal) was designed to inhibit proliferation of androgen/androgen receptor (AR)-dependent prostate cancer cell in vitro and in vivo and is currently in phase III clinical development. Additionally, clinical studies with gal revealed its superb efficacy in four different cohorts of patients with mCRPC, including those expressing splice variant AR-V7. Preclinical studies with gal show that it also exhibits strong antiproliferative activities against AR-negative prostate cancer cells and tumors through a mechanism involving phosphorylation of eIF2α, which forms an integral component of the eukaryotic mRNA translation complex. Thus, we hypothesized that gal and its new analog, VNPT55, could modulate oncogenic mRNA translation and prostate cancer cell migration and invasion. We report that gal and VNPT55 profoundly inhibit migration and invasion of prostate cancer cells, possibly by down-regulating protein expression of several EMT markers (Snail, Slug, N-cadherin, vimentin, and MMP-2/-9) via antagonizing the Mnk-eIF4E axis. In addition, gal/VNPT55 inhibited both NF-κB and Twist1 transcriptional activities, down-regulating Snail and BMI-1 mRNA expression, respectively. Furthermore, profound up-regulation of E-cadherin mRNA and protein expression may explain the observed significant inhibition of prostate cancer cell migration and invasion. Moreover, expression of self-renewal proteins, β-catenin, CD44, and Nanog, was markedly depleted. Analysis of gal/VNPT55-treated CWR22Rv1 xenograft tissue sections also revealed that observations in vitro were recapitulated in vivo. Our results suggest that gal/VNPT55 could become promising agents for the prevention and/or treatment of all stages of prostate cancer.
Collapse
Affiliation(s)
- Andrew K Kwegyir-Afful
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert D Bruno
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Francis N Murigi
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA.,Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
24
|
Storm M, Sheng X, Arnoldussen YJ, Saatcioglu F. Prostate cancer and the unfolded protein response. Oncotarget 2016; 7:54051-54066. [PMID: 27303918 PMCID: PMC5288241 DOI: 10.18632/oncotarget.9912] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/23/2016] [Indexed: 01/01/2023] Open
Abstract
The endoplasmic reticulum (ER) is an essential organelle that contributes to several key cellular functions, including lipogenesis, gluconeogenesis, calcium storage, and organelle biogenesis. The ER also serves as the major site for protein folding and trafficking, especially in specialized secretory cells. Accumulation of misfolded proteins and failure of ER adaptive capacity activates the unfolded protein response (UPR) which has been implicated in several chronic diseases, including cancer. A number of recent studies have implicated UPR in prostate cancer (PCa) and greatly expanded our understanding of this key stress signaling pathway and its regulation in PCa. Here we summarize these developments and discuss their potential therapeutic implications.
Collapse
Affiliation(s)
| | - Xia Sheng
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Yke Jildouw Arnoldussen
- Department of Biological and Chemical Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
25
|
Purushottamachar P, Kwegyir-Afful AK, Martin M, Ramamurthy V, Ramalingam S, Njar VCO. Identification of Novel Steroidal Androgen Receptor Degrading Agents Inspired by Galeterone 3β-Imidazole Carbamate. ACS Med Chem Lett 2016; 7:708-13. [PMID: 27437082 PMCID: PMC4948004 DOI: 10.1021/acsmedchemlett.6b00137] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/23/2016] [Indexed: 01/03/2023] Open
Abstract
Degradation of all forms of androgen receptors (ARs) is emerging as an advantageous therapeutic paradigm for the effective treatment of prostate cancer. In continuation of our program to identify and develop improved efficacious novel small-molecule agents designed to disrupt AR signaling through enhanced AR degradation, we have designed, synthesized, and evaluated novel C-3 modified analogues of our phase 3 clinical agent, galeterone (5). Concerns of potential in vivo stability of our recently discovered more efficacious galeterone 3β-imidazole carbamate (6) led to the design and synthesis of new steroidal compounds. Two of the 11 compounds, 3β-pyridyl ether (8) and 3β-imidazole (17) with antiproliferative GI50 values of 3.24 and 2.54 μM against CWR22Rv1 prostate cancer cell, are 2.75- and 3.5-fold superior to 5. In addition, compounds 8 and 17 possess improved (∼4-fold) AR-V7 degrading activities. Importantly, these two compounds are expected to be metabolically stable, making them suitable for further development as new therapeutics against all forms of prostate cancer.
Collapse
Affiliation(s)
- Puranik Purushottamachar
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Andrew K. Kwegyir-Afful
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Marlena
S. Martin
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Vidya
P. Ramamurthy
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Senthilmurugan Ramalingam
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Vincent C. O. Njar
- Department of Pharmacology, Center for Biomolecular Therapeutics, and Marlene Stewart
Greenbaum Cancer Center, University of Maryland
School of Medicine, 685
West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| |
Collapse
|
26
|
Stulov SV, Dugin NO, Zharkova MS, Shcherbinin DS, Kuzikov AV, Shumyantseva VV, Misharin AY, Veselovsky AV. [Interaction of novel oxazoline derivatives of 17(20)e-pregna-5,17(20)-diene with cytochrome P450 17A1]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2016; 62:38-44. [PMID: 26973185 DOI: 10.18097/pbmc20166201038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In order to find novel inhibitors of 17a-hydroxylase-17,20-lyase (cytochrome P450 17A1, CYP17A1), a key enzyme of biosynthesis of androgens, molecular docking of six new oxazoline-containing derivatives 17(20)E-pregna-5,17(20)-diene has been carried out to the active site of the crystal structure of CYP17A1 (pdb 3ruk). Results of this study indicate that: 1) complex formation of docked compounds with CYP17A1 causes their isomerization in energetically less favorable 17(20)Z-isomer; 2) the localization of the steroid moiety of all compounds in the active site is basically the same; 3) the structure of the oxazoline moiety significantly influences its position relative to heme as well as the energy of complex formation; 4) coordination of the nitrogen atom of the oxazoline moiety and the heme iron is only possible in the 17(20)Z-conformation with anti oriented double bonds 17(20), and C=N; 5) the presence of two substituents at C4' of the oxazoline moiety significantly impairs ligand binding; 6) oxazoline--and benzoxazole-containing derivatives 17(20)E-pregna-5,17(20)-diene can effectively inhibit the catalytic activity CYP17A1 and may be of interest as a basis for the development of new drugs for the treatment of androgen-dependent cancer.
Collapse
Affiliation(s)
- S V Stulov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - N O Dugin
- Institute of Biomedical Chemistry, Moscow, Russia
| | - M S Zharkova
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | - A V Kuzikov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | | |
Collapse
|
27
|
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
|
28
|
Kwegyir-Afful AK, Ramalingam S, Purushottamachar P, Ramamurthy VP, Njar VC. Galeterone and VNPT55 induce proteasomal degradation of AR/AR-V7, induce significant apoptosis via cytochrome c release and suppress growth of castration resistant prostate cancer xenografts in vivo. Oncotarget 2015; 6:27440-60. [PMID: 26196320 PMCID: PMC4695001 DOI: 10.18632/oncotarget.4578] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 07/03/2015] [Indexed: 02/04/2023] Open
Abstract
Galeterone (Gal) is a first-in-class multi-target oral small molecule that will soon enter pivotal phase III clinical trials in castration resistant prostate cancer (CRPC) patients. Gal disrupts androgen receptor (AR) signaling via inhibition of CYP17, AR antagonism and AR degradation. Resistance to current therapy is attributed to up-regulation of full-length AR (fAR), splice variants AR (AR-Vs) and AR mutations. The effects of gal and VNPT55 were analyzed on f-AR and AR-Vs (AR-V7/ARv567es) in LNCaP, CWR22Rv1 and DU145 (transfected with AR-Vs) human PC cells in vitro and CRPC tumor xenografts. Galeterone/VNPT55 decreased fAR/AR-V7 mRNA levels and implicates Mdm2/CHIP enhanced ubiquitination of posttranslational modified receptors, targeting them for proteasomal degradation. Gal and VNPT55 also induced significant apoptosis in PC cells via increased Bax/Bcl2 ratio, cytochrome-c release with concomitant cleavage of caspase 3 and PARP. More importantly, gal and VNPT55 exhibited strong in vivo anti-CRPC activities, with no apparent host toxicities. This study demonstrate that gal and VNPT55 utilize cell-based mechanisms to deplete both fAR and AR-Vs. Importantly, the preclinical activity profiles, including profound apoptotic induction and inhibition of CRPC xenografts suggest that these agents offer considerable promise as new therapeutics for patients with CRPC and those resistant to current therapy.
Collapse
Affiliation(s)
- Andrew K. Kwegyir-Afful
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Senthilmurugan Ramalingam
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Vidya P. Ramamurthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| | - Vincent C.O. Njar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
- Marlene Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201-1559, USA
| |
Collapse
|
29
|
Al-Masoudi NA, Mahdi KM, Abdul-Rida NA, Saeed BA, Engel M. A new pregnenolone analogues as privileged scaffolds in inhibition of CYP17 hydroxylase enzyme. Synthesis and in silico molecular docking study. Steroids 2015; 100:52-9. [PMID: 25988615 DOI: 10.1016/j.steroids.2015.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 04/20/2015] [Accepted: 05/07/2015] [Indexed: 10/23/2022]
Abstract
A new series of 17-(N-(arylimino)-5-pregnen-3β-ol derivatives 19-32 as well as carboxylate and acrylate analogues of pregnenolone 37-40 were synthesized and evaluated for their inhibitory activity against human CYP17 hydroxylase expressed in Escherichia coli. Compounds 32 and 37 were the most potent analogues in this series, showing inhibition activity with IC50 = 2.11 and 1.29 μM, respectively. However, the analogue 37 revealed a better selectivity profile (83.21% inhibition of hydroxylase), which is a leading candidate for further development. Molecular docking study of 37 showed binding with the amino acid residues of CYP17 through hydrogen bonds and hydrophobic interaction.
Collapse
Affiliation(s)
- Najim A Al-Masoudi
- Department of Chemistry, College of Science, University of Basrah, Basrah, Iraq; Am Tannenhof 8, 78464 Konstanz, Germany.
| | - Kuthiar M Mahdi
- Department of Chemistry, College of Education, University of Qadisiya, Qadisiya, Iraq
| | - Nabeel A Abdul-Rida
- Department of Chemistry, College of Education, University of Qadisiya, Qadisiya, Iraq
| | - Bahjat A Saeed
- Department of Chemistry, College of Education, University of Basrah, Basrah, Iraq
| | - Mathias Engel
- Institut für Pharmazeutische und Medizinische Chemie, Universität des Saarlandes, Saarbrücken, Germany
| |
Collapse
|
30
|
Iuliani M, Pantano F, Buttigliero C, Fioramonti M, Bertaglia V, Vincenzi B, Zoccoli A, Ribelli G, Tucci M, Vignani F, Berruti A, Scagliotti GV, Tonini G, Santini D. Biological and clinical effects of abiraterone on anti-resorptive and anabolic activity in bone microenvironment. Oncotarget 2015; 6:12520-8. [PMID: 25904051 PMCID: PMC4494955 DOI: 10.18632/oncotarget.3724] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/03/2015] [Indexed: 11/25/2022] Open
Abstract
Abiraterone acetate (ABI) is associated not only with a significant survival advantage in both chemotherapy-naive and -treated patients with metastatic castration-resistant prostate cancer (mCRPC), but also with a delay in time to development of Skeletal Related Events and in radiological skeletal progression. These bone benefits may be related to a direct effect on prostate cancer cells in bone or to a specific mechanism directed to bone microenvironment. To test this hypothesis we designed an in vitro study aimed to evaluate a potential direct effect of ABI on human primary osteoclasts/osteoblasts (OCLs/OBLs). We also assessed changes in bone turnover markers, serum carboxy-terminal collagen crosslinks (CTX) and alkaline phosphatase (ALP), in 49 mCRPC patients treated with ABI.Our results showed that non-cytotoxic doses of ABI have a statistically significant inhibitory effect on OCL differentiation and activity inducing a down-modulation of OCL marker genes TRAP, cathepsin K and metalloproteinase-9. Furthermore ABI promoted OBL differentiation and bone matrix deposition up-regulating OBL specific genes, ALP and osteocalcin. Finally, we observed a significant decrease of serum CTX values and an increase of ALP in ABI-treated patients.These findings suggest a novel biological mechanism of action of ABI consisting in a direct bone anabolic and anti-resorptive activity.
Collapse
Affiliation(s)
- Michele Iuliani
- Translational Oncology Laboratory, Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Francesco Pantano
- Translational Oncology Laboratory, Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Consuelo Buttigliero
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Marco Fioramonti
- Translational Oncology Laboratory, Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Valentina Bertaglia
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Bruno Vincenzi
- Translational Oncology Laboratory, Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Alice Zoccoli
- Translational Oncology Laboratory, Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Giulia Ribelli
- Translational Oncology Laboratory, Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Marcello Tucci
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Francesca Vignani
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Alfredo Berruti
- U.O. Oncologia Medica, Ospedali Civili di Brescia, Brescia, Italy
| | | | - Giuseppe Tonini
- Translational Oncology Laboratory, Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| | - Daniele Santini
- Translational Oncology Laboratory, Medical Oncology, University Campus Bio-Medico of Rome, Rome, Italy
| |
Collapse
|
31
|
Gomez L, Kovac JR, Lamb DJ. CYP17A1 inhibitors in castration-resistant prostate cancer. Steroids 2015; 95:80-7. [PMID: 25560485 PMCID: PMC4323677 DOI: 10.1016/j.steroids.2014.12.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/14/2014] [Accepted: 12/22/2014] [Indexed: 10/24/2022]
Abstract
The majority of prostate cancer (PCa) cases are diagnosed as a localized disease. Definitive treatment, active surveillance or watchful waiting are employed as therapeutic paradigms. The current standard of care for the treatment of metastatic PCa is either medical or surgical castration. Once PCa progresses in spite of castrate androgen levels it is termed 'castration-resistant prostate cancer' (CRPC). Patients may even exhibit rising PSA levels with possible bone, lymph node or solid organ metastases. In 2010, the only agent approved for the treatment of CRPC was docetaxel, a chemotherapeutic agent. It is now known that cells from patients with CRPC express androgen receptors (AR) and remain continuously influenced by androgens. As such, treatments with novel hormonal agents that specifically target the biochemical conversion of cholesterol to testosterone have come to the forefront. The use of cytochrome P450c17 (CYP17A1) inhibitor underlies one of the most recent advances in the treatment of CRPC. Abiraterone acetate (AA) was the first CYP17A1 inhibitor approved in the United States. This review will discuss CRPC in general with a specific focus on AA and novel CYP17A1 inhibitors. AA clinical trials will be reviewed along with other novel adjunct treatments that may enhance the effectiveness of abiraterone therapy. Furthermore, the most recently identified CYP17A1 inhibitors Orteronel, Galeterone, VT-464, and CFG920 will also be explored.
Collapse
Affiliation(s)
- Lissette Gomez
- Scott Department of Urology and The Center for Reproductive Medicine, and the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Jason R Kovac
- Urology of Indiana, Male Reproductive Endocrinology and Surgery, Carmel, IN, United States
| | - Dolores J Lamb
- Scott Department of Urology and The Center for Reproductive Medicine, and the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
32
|
Njar VCO, Brodie AMH. Discovery and Development of Galeterone (TOK-001 or VN/124-1) for the Treatment of All Stages of Prostate Cancer. J Med Chem 2015; 58:2077-87. [DOI: 10.1021/jm501239f] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Vincent C. O. Njar
- Department of Pharmacology, ‡Center for Biomolecular
Therapeutics, and §Marlene Stewart
Greenebaum Cancer Center, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | - Angela M. H. Brodie
- Department of Pharmacology, ‡Center for Biomolecular
Therapeutics, and §Marlene Stewart
Greenebaum Cancer Center, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| |
Collapse
|
33
|
Abstract
Prostate cancer is the second leading cause of death in adult males in the USA. Recent advances have revealed that the fatal form of this cancer, known as castration-resistant prostate cancer (CRPC), remains hormonally driven despite castrate levels of circulating androgens. CRPC arises as the tumor undergoes adaptation to low levels of androgens by either synthesizing its own androgens (intratumoral androgens) or altering the androgen receptor (AR). This article reviews the major routes to testosterone and dihydrotestosterone synthesis in CRPC cells and examines the enzyme targets and progress in the development of isoform-specific inhibitors that could block intratumoral androgen biosynthesis. Because redundancy exists in these pathways, it is likely that inhibition of a single pathway will lead to upregulation of another so that drug resistance would be anticipated. Drugs that target multiple pathways or bifunctional agents that block intratumoral androgen biosynthesis and antagonize the AR offer the most promise. Optimal use of enzyme inhibitors or AR antagonists to ensure maximal benefits to CRPC patients will also require application of precision molecular medicine to determine whether a tumor in a particular patient will be responsive to these treatments either alone or in combination.
Collapse
Affiliation(s)
- Trevor M Penning
- Perelman School of MedicineCenter of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, USA
| |
Collapse
|
34
|
Yu Z, Cai C, Gao S, Simon NI, Shen HC, Balk SP. Galeterone prevents androgen receptor binding to chromatin and enhances degradation of mutant androgen receptor. Clin Cancer Res 2014; 20:4075-85. [PMID: 24874833 DOI: 10.1158/1078-0432.ccr-14-0292] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Galeterone inhibits the enzyme CYP17A1 and is currently in phase II clinical trials for castration-resistant prostate cancer (CRPC). Galeterone is also a direct androgen receptor (AR) antagonist and may enhance AR degradation. This study was undertaken to determine the molecular basis for AR effects and their therapeutic potential. EXPERIMENTAL DESIGN Effects of galeterone on AR expression and activities were examined in prostate cancer cell lines. RESULTS Similar to the AR antagonist enzalutamide, but in contrast to bicalutamide, galeterone did not induce binding of a constitutively active VP16-AR fusion protein to reporter genes and did not induce AR recruitment to endogenous androgen-regulated genes based on chromatin immunoprecipitation. Galeterone at low micromolar concentrations that did not induce cellular stress responses enhanced AR protein degradation in LNCaP and C4-2 cells, which express a T878A mutant AR, but not in prostate cancer cells expressing wild-type AR. Further transfection studies using stable LNCaP and PC3 cell lines ectopically expressing wild-type or T878A-mutant ARs confirmed that galeterone selectively enhances degradation of the T878A-mutant AR. CONCLUSIONS Similar to enzalutamide, galeterone may be effective as a direct AR antagonist in CRPC. It may be particularly effective against prostate cancer cells with the T878A AR mutation but may also enhance degradation of wild-type AR in vivo through a combination of direct and indirect mechanisms. Finally, these findings show that conformational changes in AR can markedly enhance its degradation and thereby support efforts to develop further antagonists that enhance AR degradation.
Collapse
Affiliation(s)
- Ziyang Yu
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Changmeng Cai
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Shuai Gao
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Nicholas I Simon
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Howard C Shen
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Steven P Balk
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Abstract
The discovery of androgen dependence in prostate cancer in 1941 by Huggins and colleagues has remained the backbone for the treatment of this disease. However, although many patients initially respond to androgen depletion therapy, they almost invariably relapse and develop resistance with transition of the disease to a castration-resistant state. Over the past decade, the better understanding of the mechanisms that drive resistance to castration has led to the development of next-generation androgen receptor targeting agents such as abiraterone acetate and enzalutamide. This Review aims to revisit the discovery and evolution of androgen receptor targeting therapeutics for the treatment of advanced-stage prostate cancer over the years and to discuss the upcoming future and challenges in the treatment of this common cancer.
Collapse
|
36
|
Godbole AM, Ramalingam S, Ramamurthy VP, Khandelwal A, Bruno RD, Upreti VV, Gediya LK, Purushottamachar P, Mbatia HW, Addya S, Ambulos N, Njar VCO. VN/14-1 induces ER stress and autophagy in HP-LTLC human breast cancer cells and has excellent oral pharmacokinetic profile in female Sprague Dawley rats. Eur J Pharmacol 2014; 734:98-104. [PMID: 24726842 DOI: 10.1016/j.ejphar.2014.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 12/19/2022]
Abstract
Resistance to aromatase inhibitors is a major concern in the treatment of breast cancer. Long-term letrozole cultured (LTLC) cells represent a model of resistance to aromatase inhibitors. The LTLC cells were earlier generated by culturing MCF-7Ca, the MCF-7 human breast cancer cell line stably transfected with human placental aromatase gene for a prolonged period in the presence of letrozole. In the present study the effect of RAMBA, VN/14-1 on the sensitivity of LTLC cells upon multiple passaging and the mechanisms of action of VN/14-1 in such high passage LTLC (HP-LTLC) cells was investigated. We report that multiple passaging of LTLC cells (HP-LTLC cell clones) led to profound decrease in their sensitivity to VN/14-1. Additionally, microarray studies and protein analysis revealed that VN/14-1 induced marked endoplasmic reticulum (ER) stress and autophagy in HP-LTLC cells. We further report that VN/14-1 in combination with thapsigargin exhibited synergistic anti-cancer effect in HP-LTLC cells. Preliminary pharmacokinetics in rats revealed that VN/14-1 reached a peak plasma concentration (Cmax) within 0.17h after oral dosing. Its absolute oral bioavailability was >100%. Overall these results indicate potential of VN/14-1 for further clinical development as a potential oral agent for the treatment of breast cancer.
Collapse
Affiliation(s)
- Abhijit M Godbole
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Senthilmurugan Ramalingam
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vidya P Ramamurthy
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Aakanksha Khandelwal
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Robert D Bruno
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vijay V Upreti
- Department of Pharmaceutical sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
| | - Lalji K Gediya
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Hannah W Mbatia
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Sankar Addya
- Department of Cancer Biology, Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, PA 19107, USA
| | - Nicholas Ambulos
- Genomic Core Facility, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Marlene Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA.
| |
Collapse
|
37
|
Kotovshchikov YN, Latyshev GV, Lukashev NV, Beletskaya IP. Synthesis of novel 1,2,3-triazolyl derivatives of pregnane, androstane andd-homoandrostane. Tandem “click” reaction/Cu-catalyzedd-homo rearrangement. Org Biomol Chem 2014; 12:3707-20. [DOI: 10.1039/c4ob00404c] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
38
|
Neuzillet Y, Flamand V, Lebdai S, Villers A, Lebret T. [Prostate cancer and new hormonal treatments: mechanism of action and main clinical results]. Prog Urol 2013; 23 Suppl 1:S34-43. [PMID: 24314737 DOI: 10.1016/s1166-7087(13)70044-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION New drugs have recently been developed, through a better understanding of the mechanisms involved in the progression of prostate cancer, including castration-resistant ones (CRPC). This article aims to describe the mechanisms of action of these new hormonal treatments and their major clinical outcomes and development programs. MATERIALS AND METHODS A bibliographic research in French and English using Medline(®) and Embase(®) using the keywords "castration-resistant prostate cancer", "abiraterone acetate", "orteronel", "enzalutamide", and "clinical trials" was performed. RESULTS the androgen signaling pathway remains the cornerstone of advanced cancers management. Hence, some molecules target the androgen biosynthesis, as abiraterone acetate and orteronel, which are selective inhibitors of the enzyme CYP17. Others act as antagonists of the androgen receptor: the enzalutamide, RNA-509 and ODM201. Finally, galeterone combines the two effects. CONCLUSION Progress conferred by these molecules in terms of overall survival and quality of life in patients with metastatic CRPC, suggest that their use at earlier stages of the disease could reduce morbidity and mortality from prostate cancer. Determining the best strategy for sequence or combination therapy to optimize the use of these new molecules should be investigated.
Collapse
Affiliation(s)
- Y Neuzillet
- Service d'urologie, hôpital Foch, université de Versailles - Saint-Quentin-en-Yvelines (UVSQ), 92150 Suresnes, France.
| | | | | | | | | |
Collapse
|
39
|
Van Hemelrijck M, Michaelsson K, Nelson WG, Kanarek N, Dobbs A, Platz EA, Rohrmann S. Association of serum calcium with serum sex steroid hormones in men in NHANES III. Aging Male 2013; 16:151-8. [PMID: 23672276 PMCID: PMC4005327 DOI: 10.3109/13685538.2013.772133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Bone is a positive regulator of male fertility, which indicates a link between regulation of bone remodeling and reproduction or more specifically a link between calcium and androgens. This possibly suggests how calcium is linked to prostate cancer development through its link with the reproductive system. We studied serum calcium and sex steroid hormones in the Third National Health and Nutrition Examination Survey (NHANES III). METHODS Serum calcium and sex steroid hormones were measured for 1262 men in NHANES III. We calculated multivariable-adjusted geometric means of serum concentrations of total and estimated free testosterone and estradiol, androstanediol glucuronide (AAG), and sex hormone binding globulin (SHBG) by categories of calcium (lowest 5% [<1.16 mmol/L], mid 90%, top 5% [≥1.30 mmol/L]). RESULTS Levels of total and free testosterone, total estradiol or AAG did not differ across categories of serum calcium. Adjusted SHBG concentrations were 36.4 for the bottom 5%, 34.2 for the mid 90% and 38.9 nmol/L for the top 5% of serum calcium (Ptrend = 0.006), free estradiol levels were 0.88, 0.92 and 0.80 pg/ml (Ptrend = 0.048). CONCLUSIONS This link between calcium and sex steroid hormones, in particular the U-shaped pattern with SHBG, may, in part, explain why observational studies have found a link between serum calcium and risk of prostate cancer.
Collapse
Affiliation(s)
- Mieke Van Hemelrijck
- King’s College London, School of Medicine, Division of Cancer Studies, Cancer Epidemiology Unit, London, UK
| | - Karl Michaelsson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - William G Nelson
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
- James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Departments of Oncology, Pathology, Pharmacology and Molecular Sciences, Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Norma Kanarek
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Adrian Dobbs
- Johns Hopkins University School of Medicine, Division of Endocrinology and Metabolism, Baltimore, MD, USA
| | - Elizabeth A Platz
- Johns Hopkins Bloomberg School of Public Health, Department of Epidemiology, Baltimore, MD, USA
| | - Sabine Rohrmann
- University of Zurich, Institute of Social and Preventive Medicine, Zurich, Switzerland
| |
Collapse
|
40
|
17(E)-Picolinylidene androstane derivatives as potential inhibitors of prostate cancer cell growth: Antiproliferative activity and molecular docking studies. Bioorg Med Chem 2013; 21:7257-66. [DOI: 10.1016/j.bmc.2013.09.063] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 11/17/2022]
|
41
|
Abstract
As the first in class steroid 17α-hydroxylase/C17,20-lyase (CYP17) inhibitor, abiraterone acetate (of which the active metabolite is abiraterone) has been shown to improve overall survival in patients with castration-resistant prostate cancer (CRPC)--in those who are chemotherapy-naive and those previously treated with docetaxel. Furthermore, the clinical success of abiraterone demonstrated that CRPC, which has previously been regarded as an androgen-independent disease, is still driven, at least in part, by androgens. More importantly, abiraterone is a 'promiscuous' drug that interacts with a number of targets, which dictate its clinical benefits and adverse effects profile. Besides CYP17 inhibition, abiraterone acts as an antagonist to the androgen receptor and inhibits 3β-hydroxysteroid dehydrogenase--two effects that potentially contribute to its antitumour effects. However, the inhibition of the 17α-hydroxylase activity of CYP17, CYP11B1 and a panel of hepatic CYP enzymes leads to adverse effects and toxicities that include secondary mineralocorticoid excess. Abiraterone is also associated with increased incidence of cardiac disorders. Under such circumstances, development of new CYP17 inhibitors as an additional line of defence is urgently needed. To achieve enhanced clinical benefits, new strategies are being explored that include selective inhibition of the C17,20-lyase activity of CYP17 and multi-targeting strategies that affect androgen synthesis and signalling at different points. Some of these strategies-including the drugs orteronel, VT-464 and galeterone--are supported by preclinical data and are being explored in the clinic.
Collapse
|
42
|
Heidenreich A, Pfister D, Merseburger A, Bartsch G. Castration-resistant Prostate Cancer: Where We Stand in 2013 and What Urologists Should Know. Eur Urol 2013; 64:260-5. [DOI: 10.1016/j.eururo.2013.05.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/05/2013] [Indexed: 10/26/2022]
|
43
|
Abstract
In April 2011, abiraterone acetate (in combination with low-dose steroids) was approved by the US Food and Drug Administration for the treatment of men with metastatic, castration-resistant prostate cancer who have previously been treated with docetaxel-based chemotherapy. The development of abiraterone was the successful result of an improved understanding of the role of the androgen receptor signaling pathway in castration-resistant prostate cancer. Abiraterone is a rationally designed potent inhibitor of cytochrome P450, family 17, subfamily A, polypeptide 1, which is essential for synthesis of testosterone from nongonadal precursors. More recently, other drugs that act along the androgen0synthesis pathway, such as orteronel (TAK-700) and galeterone (TOK-001), have shown promise in early clinical trials. Here, we review the discovery and clinical development of abiraterone and other novel androgen-synthesis inhibitors for the management of advanced prostate cancer.
Collapse
|
44
|
Lallous N, Dalal K, Cherkasov A, Rennie PS. Targeting alternative sites on the androgen receptor to treat castration-resistant prostate cancer. Int J Mol Sci 2013; 14:12496-519. [PMID: 23771019 PMCID: PMC3709796 DOI: 10.3390/ijms140612496] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 05/29/2013] [Accepted: 06/03/2013] [Indexed: 01/22/2023] Open
Abstract
Recurrent, metastatic prostate cancer continues to be a leading cause of cancer-death in men. The androgen receptor (AR) is a modular, ligand-inducible transcription factor that regulates the expression of genes that can drive the progression of this disease, and as a consequence, this receptor is a key therapeutic target for controlling prostate cancer. The current drugs designed to directly inhibit the AR are called anti-androgens, and all act by competing with androgens for binding to the androgen/ligand binding site. Unfortunately, with the inevitable progression of the cancer to castration resistance, many of these drugs become ineffective. However, there are numerous other regulatory sites on this protein that have not been exploited therapeutically. The regulation of AR activity involves a cascade of complex interactions with numerous chaperones, co-factors and co-regulatory proteins, leading ultimately to direct binding of AR dimers to specific DNA androgen response elements within the promoter and enhancers of androgen-regulated genes. As part of the family of nuclear receptors, the AR is organized into modular structural and functional domains with specialized roles in facilitating their inter-molecular interactions. These regions of the AR present attractive, yet largely unexploited, drug target sites for reducing or eliminating androgen signaling in prostate cancers. The design of small molecule inhibitors targeting these specific AR domains is only now being realized and is the culmination of decades of work, including crystallographic and biochemistry approaches to map the shape and accessibility of the AR surfaces and cavities. Here, we review the structure of the AR protein and describe recent advancements in inhibiting its activity with small molecules specifically designed to target areas distinct from the receptor’s androgen binding site. It is anticipated that these new classes of anti-AR drugs will provide an additional arsenal to treat castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Nada Lallous
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada.
| | | | | | | |
Collapse
|
45
|
Ferraldeschi R, Pezaro C, Karavasilis V, de Bono J. Abiraterone and Novel Antiandrogens: Overcoming Castration Resistance in Prostate Cancer. Annu Rev Med 2013; 64:1-13. [DOI: 10.1146/annurev-med-121211-091605] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- R. Ferraldeschi
- Division of Cancer Therapeutics, Signal Transduction & Molecular Pharmacology and Clinical Pharmacology & Trials Team, Institute of Cancer Research, Sutton, SM25NG, United Kingdom;
| | - C. Pezaro
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Sutton, SM25PT, United Kingdom; ,
| | - V. Karavasilis
- Medical Oncology Department, Aristotle University of Thessaloniki, Thessaloniki, Greece;
| | - J. de Bono
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Sutton, SM25PT, United Kingdom; ,
| |
Collapse
|
46
|
Godbole AM, Purushottamachar P, Martin MS, Daskalakis C, Njar VCO. Autophagy inhibition synergistically enhances anticancer efficacy of RAMBA, VN/12-1 in SKBR-3 cells, and tumor xenografts. Mol Cancer Ther 2012; 11:898-908. [PMID: 22334589 DOI: 10.1158/1535-7163.mct-11-0860] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
VN/12-1 is a novel retinoic acid metabolism blocking agent discovered in our laboratory. The purpose of the study was to elucidate the molecular mechanism of anticancer activity of VN/12-1 in breast cancer cell lines and in tumor xenografts. We investigated the effects of VN/12-1 on induction of autophagy and apoptosis in SKBR-3 cells. Furthermore, we also examined the impact of pharmacologic and genomic inhibition of autophagy on anticancer activity of VN/12-1. Finally, the antitumor activity of VN/12-1 was evaluated as a single agent and in combination with autophagy inhibitor chloroquine in an SKBR-3 mouse xenograft model. Short exposure of low dose (<10 μmol/L) of VN/12-1 induced endoplasmic reticulum stress, autophagy, and inhibited G(1)-S phase transition and caused a protective response. However, a higher dose of VN/12-1 initiated apoptosis in vitro. Inhibition of autophagy using either pharmacologic inhibitors or RNA interference of Beclin-1 enhanced anticancer activity induced by VN/12-1 in SKBR-3 cells by triggering apoptosis. Importantly, VN/12-1 (5 mg/kg twice weekly) and the combination of VN/12-1 (5 mg/kg twice weekly) + chloroquine (50 mg/kg twice weekly) significantly suppressed established SKBR-3 tumor growth by 81.4% (P < 0.001 vs. control) and 96.2% (P < 0.001 vs. control), respectively. Our novel findings suggest that VN/12-1 may be useful as a single agent or in combination with autophagy inhibitors for treating human breast cancers. Our data provides a strong rationale for clinical evaluation of VN/12-1 as single agent or in combination with autophagy inhibitors.
Collapse
Affiliation(s)
- Abhijit M Godbole
- Department of Pharmaceutical Sciences, Jefferson School of Pharmacy, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
47
|
Soifer HS, Souleimanian N, Wu S, Voskresenskiy AM, Collak FK, Cinar B, Stein CA. Direct regulation of androgen receptor activity by potent CYP17 inhibitors in prostate cancer cells. J Biol Chem 2011; 287:3777-87. [PMID: 22174412 DOI: 10.1074/jbc.m111.261933] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
TOK-001 and abiraterone are potent 17-heteroarylsteroid (17-HAS) inhibitors of Cyp17, one of the rate-limiting enzymes in the biosynthesis of testosterone from cholesterol in prostate cancer cells. Nevertheless, the molecular mechanism underlying the prevention of prostate cell growth by 17-HASs still remains elusive. Here, we assess the effects of 17-HASs on androgen receptor (AR) activity in LNCaP and LAPC-4 cells. We demonstrate that both TOK-001 and abiraterone reduced AR protein and mRNA expression, and antagonized AR-dependent promoter activation induced by androgen. TOK-001, but not abiraterone, is an effective apparent competitor of the radioligand [(3)H]R1881 for binding to the wild type and various mutant AR (W741C, W741L) proteins. In agreement with these data, TOK-001 is a consistently superior inhibitor than abiraterone of R1881-induced transcriptional activity of both wild type and mutant AR. However, neither agent was able to trans-activate the AR in the absence of R1881. Our data demonstrate that phospho-4EBP1 levels are significantly reduced by TOK-001 and to a lesser extent by abiraterone alcohol, and suggest a mechanism by which cap-dependent translation is suppressed by blocking assembly of the eIF4F and eIF4G complex to the mRNA 5' cap. Thus, the effects of these 17-HASs on AR signaling are complex, ranging from a decrease in testosterone production through the inhibition of Cyp17 as previously described, to directly reducing both AR protein expression and R1881-induced AR trans-activation.
Collapse
|
48
|
Synthesis of novel 4-(1H-benzimidazol-2-yl)benzene-1,3-diols and their cytotoxic activity against human cancer cell lines. Arch Pharm Res 2011; 34:1639-47. [DOI: 10.1007/s12272-011-1008-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/16/2011] [Accepted: 06/27/2011] [Indexed: 10/15/2022]
|
49
|
Mostaghel EA, Plymate S. New hormonal therapies for castration-resistant prostate cancer. Endocrinol Metab Clin North Am 2011; 40:625-42, x. [PMID: 21889725 PMCID: PMC3167094 DOI: 10.1016/j.ecl.2011.05.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Continued activation of the androgen receptor (AR) axis despite castration remains a critical force in the development of castration-resistant prostate cancer (CRPC). Therapeutic strategies designed to more effectively ablate tumoral androgen activity are required to improve clinical efficacy and prevent disease progression. Tumor-based alterations in expression and activity of the AR and in steroidogenic pathways mediating ligand generation facilitate the development of CRPC. This article reviews AR and ligand-dependent mechanisms underlying CRPC progression and the status of novel hormonal therapies targeting the AR axis that are currently in clinical and preclinical development.
Collapse
Affiliation(s)
- Elahe A Mostaghel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | |
Collapse
|
50
|
Advances in small molecule inhibitors of androgen receptor for the treatment of advanced prostate cancer. World J Urol 2011; 30:311-8. [PMID: 21833557 DOI: 10.1007/s00345-011-0745-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 07/29/2011] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Current treatments for localized prostate cancer include brachytherapy, external beam radiation, surgery, and active surveillance. Unfortunately, 20-40% of prostate cancer patients will experience recurrence and require hormonal therapies. These therapies involve androgen ablation by chemical or surgical castration and application of antiandrogens. Hormonal therapy is initially effective, but will inevitably fail and the disease will progress to lethal castration-resistant prostate cancer (CRPC) from which patients succumb within 2 years. CRPC is considered to be dependent on transcriptionally active androgen receptors (AR). This article reviews recent advances in the discovery and development of small molecule inhibitors of AR. METHODS A PubMed database search was performed for articles focused on small molecule inhibitors of AR for potential development for the treatment of prostate cancer. Compounds with broad effects on other pathways were not included. RESULTS Currently, there are several novel antiandrogens being tested in the clinic that have improved affinity for the AR and work by different mechanisms to the current battery of approved antiandrogens that are discussed. Small molecule inhibitors that interact with regions other than the AR ligand-binding pocket have been also been discovered. These small molecules include allosteric inhibitors of the LBD, compounds that alter AR conformation, and antagonists to the AR NTD and are highlighted. CONCLUSIONS CRPC is dependent upon transcriptionally active AR. Survival improvement may be achieved by complete blockade of all AR activity using novel small molecule inhibitors with unique mechanisms of action.
Collapse
|