1
|
Cheng Y, Yu TT, Olzomer EM, Beretta M, Katen A, Su J, Jones JP, Black DS, Hoehn KL, Byrne FL, Kumar N. Design, synthesis and biological evaluation of naphthalene-1,4-dione analogues as anticancer agents. RSC Med Chem 2025:d4md00987h. [PMID: 40223824 PMCID: PMC11986672 DOI: 10.1039/d4md00987h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
The increased metabolism of glucose via aerobic glycolysis, known as the Warburg effect, is a hallmark of most cancers. Identifying molecules that disrupt the Warburg effect may allow for selective cytotoxicity towards cancer cells and reduce side effects compared to current chemotherapy agents. Our initial hit compound, BH10, which potentially targets Kelch-like ECH-associated protein 1 (Keap1), increased oxygen consumption rate and displayed increased cytotoxicity towards cancer cells over normal cells in vitro. In this project, a library of analogues based on the BH10 scaffold was prepared with the aim of improving potency and cancer-cell specificity. Among these analogues, several compounds showed notable potency, with activity (IC50) observed around 1 μM. However, when considering selectivity, the imidazole derivative, compound 44, exhibited the most optimal balance, achieving an IC50 of 6.4 μM and selectivity ratio of 3.6 which indicates greater toxicity to cancer cells vs. normal cells.
Collapse
Affiliation(s)
- Yao Cheng
- School of Chemistry, University of New South Wales Sydney NSW 2052 Australia
| | - Tsz Tin Yu
- School of Chemistry, University of New South Wales Sydney NSW 2052 Australia
| | - Ellen M Olzomer
- School of Biotechnology and Biomolecular Sciences, University of New South Wales Sydney NSW 2052 Australia
| | - Martina Beretta
- School of Biotechnology and Biomolecular Sciences, University of New South Wales Sydney NSW 2052 Australia
| | - Alice Katen
- School of Chemistry, University of New South Wales Sydney NSW 2052 Australia
| | - Jacky Su
- School of Chemistry, University of New South Wales Sydney NSW 2052 Australia
| | - John Patrick Jones
- School of Chemistry, University of New South Wales Sydney NSW 2052 Australia
| | - David StC Black
- School of Chemistry, University of New South Wales Sydney NSW 2052 Australia
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales Sydney NSW 2052 Australia
| | - Frances L Byrne
- School of Biotechnology and Biomolecular Sciences, University of New South Wales Sydney NSW 2052 Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales Sydney NSW 2052 Australia
| |
Collapse
|
2
|
Yuan J, Liu Z, Dong Y, Gao F, Xia X, Wang P, Luo Y, Zhang Z, Yan D, Zhang W. Pioneering 4,11-Dioxo-4,11-dihydro-1 H-anthra[2,3- d]imidazol-3-ium Compounds as Promising Survivin Inhibitors by Targeting ILF3/NF110 for Cancer Therapy. J Med Chem 2023; 66:16843-16868. [PMID: 38079530 DOI: 10.1021/acs.jmedchem.3c01551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Survivin is a novel attractive target for cancer therapy; however, it is considered undruggable because it lacks enzymatic activities. Herein, we describe our efforts toward the discovery of a novel series of 4,11-dioxo-4,11-dihydro-1H-anthra[2,3-d]imidazol-3-ium derivatives as survivin inhibitors by targeting ILF3/NF110. Intensive structural modifications led us to identify a lead compound AQIM-I, which remarkably inhibited nonsmall cell lung cancer cells A549 with an IC50 value of 9 nM and solid tumor cell proliferation with more than 700-fold selectivity against human normal cells. Further biological studies revealed that compound AQIM-I significantly inhibited survivin expression and colony formation and induced ROS production, apoptosis, cell cycle arrest, DNA damage, and autophagy. Furthermore, the promoter-luciferase reporter assay showed that AQIM-I attenuated the survivin promoter activity enhanced by the overexpression of ILF3/NF110 in a concentration-dependent manner, and specific binding (KD = 163 nM) of AQIM-I to ILF3/NF110 was detected by surface plasmon resonance.
Collapse
Affiliation(s)
- Jing Yuan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Zhanxiong Liu
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Yachun Dong
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Feng Gao
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Xuelin Xia
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Penghui Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Yanli Luo
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P. R. China
| | - Zhenfeng Zhang
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Deyue Yan
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Wanbin Zhang
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| |
Collapse
|
3
|
Cui Q, Huang C, Liu JY, Zhang JT. Small Molecule Inhibitors Targeting the "Undruggable" Survivin: The Past, Present, and Future from a Medicinal Chemist's Perspective. J Med Chem 2023; 66:16515-16545. [PMID: 38092421 PMCID: PMC11588358 DOI: 10.1021/acs.jmedchem.3c01130] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Survivin, a homodimeric protein and a member of the IAP family, plays a vital function in cell survival and cycle progression by interacting with various proteins and complexes. Its expression is upregulated in cancers but not detectable in normal tissues. Thus, it has been regarded and validated as an ideal cancer target. However, survivin is "undruggable" due to its lack of enzymatic activities or active sites for small molecules to bind/inhibit. Academic and industrial laboratories have explored different strategies to overcome this hurdle over the past two decades, with some compounds advanced into clinical testing. These strategies include inhibiting survivin expression, its interaction with binding partners and homodimerization. Here, we provide comprehensive analyses of these strategies and perspective on different small molecule survivin inhibitors to help drug discovery targeting "undruggable" proteins in general and survivin specifically with a true survivin inhibitor that will prevail in the foreseeable future.
Collapse
Affiliation(s)
- Qingbin Cui
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Caoqinglong Huang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jing-Yuan Liu
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| |
Collapse
|
4
|
Chen GQ, Guo HY, Quan ZS, Shen QK, Li X, Luan T. Natural Products-Pyrazine Hybrids: A Review of Developments in Medicinal Chemistry. Molecules 2023; 28:7440. [PMID: 37959859 PMCID: PMC10649211 DOI: 10.3390/molecules28217440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Pyrazine is a six-membered heterocyclic ring containing nitrogen, and many of its derivatives are biologically active compounds. References have been downloaded through Web of Science, PubMed, Science Direct, and SciFinder Scholar. The structure, biological activity, and mechanism of natural product derivatives containing pyrazine fragments reported from 2000 to September 2023 were reviewed. Publications reporting only the chemistry of pyrazine derivatives are beyond the scope of this review and have not been included. The results of research work show that pyrazine-modified natural product derivatives have a wide range of biological activities, including anti-inflammatory, anticancer, antibacterial, antiparasitic, and antioxidant activities. Many of these derivatives exhibit stronger pharmacodynamic activity and less toxicity than their parent compounds. This review has a certain reference value for the development of heterocyclic compounds, especially pyrazine natural product derivatives.
Collapse
Affiliation(s)
- Guo-Qing Chen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Hong-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Qing-Kun Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Xiaoting Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Tian Luan
- Department of Pharmacy, Shenyang Medical College, Shenyang 110034, China
| |
Collapse
|
5
|
Devi M, Kumar P, Singh R, Narayan L, Kumar A, Sindhu J, Lal S, Hussain K, Singh D. A comprehensive review on synthesis, biological profile and photophysical studies of heterocyclic compounds derived from 2,3-diamino-1,4-naphthoquinone. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6
|
Wang X, Cheng Y, Yuan Y, Zhang Y, Wang W. Structures and electron affinity energies of polycyclic quinones. Heliyon 2022; 8:e10107. [PMID: 35991986 PMCID: PMC9389180 DOI: 10.1016/j.heliyon.2022.e10107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/09/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
In this study, quinoid structures, semiquinone radical structures, and electron affinity energies (EAEs) of many polycyclic quinones containing heteroatoms (O, B, and F) or heterocycles (pyrrole, imidazole, and pyrazine) were calculated. Quinones with unstable quinoid structures and stable semiquinone radical structures had high EAEs. The main factors of quinoid structural instability were spatial repulsion and antiaromaticity, and the stability factors of the semiquinone radical structure comprised inductive effects, hydrogen bonds, electrostatic interactions, and orbital interactions. Compound 11 had both the antiaromaticity of the quinoid structure and the orbital interactions of the semiquinone radical structure, thus having the highest EAE. The crystal structure of compound 8 was obtained, and it confirmed the reliability of the calculated results of this work. The relationship between electron affinity energy and many factors is revealed. The close relationship between the electronic structure and the spatial structure is revealed. Various interactions such as orbital, electrostatic, and spatial repulsion are exhibited.
Collapse
|
7
|
Mackay RP, Weinberger PM, Copland JA, Mahdavian E, Xu Q. YM155 Induces DNA Damage and Cell Death in Anaplastic Thyroid Cancer Cells by Inhibiting DNA Topoisomerase IIα at the ATP-Binding Site. Mol Cancer Ther 2022; 21:925-935. [PMID: 35405742 PMCID: PMC9167740 DOI: 10.1158/1535-7163.mct-21-0619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 12/10/2021] [Accepted: 04/01/2022] [Indexed: 11/16/2022]
Abstract
Anaplastic thyroid cancer (ATC) is among the most aggressive of human cancers, and currently there are few effective treatments for most patients. YM155, first identified as a survivin inhibitor, was highlighted in a high-throughput screen performed by the National Cancer Institute, killing ATC cells in vitro and in vivo. However, there was no association between survivin expression and response to YM155 in clinical trials, and YM155 has been mostly abandoned for development despite favorable pharmacokinetic and toxicity profiles. Currently, alternative mechanisms are being explored for YM155 by a number of groups. In this study, ATC patient samples show overexpression of topoisomerase Top2α compared with benign thyroid samples and to differentiated thyroid cancers. ATC cell lines that overexpress Top2α are more sensitive to YM155. We created a YM155-resistant cell line, which shows decreased expression of Top2α and is resensitized with Top2α overexpression. Molecular modeling predicts binding for YM155 in the Top2α ATP-binding site and identifies key amino acids for YM155-Top2α interaction. A Top2α mutant abrogates the effect of YM155, confirming the contribution of Top2α to YM155 mechanism of action. Our results suggest a novel mechanism of action for YM155 and may represent a new therapeutic approach for the treatment of ATC.
Collapse
Affiliation(s)
- Ryan P. Mackay
- Department of Otolaryngology-Head & Neck Surgery, Louisiana State University Health Sciences Center – Shreveport, Shreveport, LA, United States
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, United States
| | - Paul M. Weinberger
- Department of Otolaryngology-Head & Neck Surgery, Louisiana State University Health Sciences Center – Shreveport, Shreveport, LA, United States
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, United States
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Elahe Mahdavian
- Department of Biological Sciences, Louisiana State University in Shreveport, Shreveport, LA, United States
| | - Qinqin Xu
- Department of Otolaryngology-Head & Neck Surgery, Louisiana State University Health Sciences Center – Shreveport, Shreveport, LA, United States
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, United States
| |
Collapse
|
8
|
Tabassum R, Ashfaq M, Oku H. Current Pharmaceutical Aspects of Synthetic Quinoline Derivatives. Mini Rev Med Chem 2021; 21:1152-1172. [PMID: 33319670 DOI: 10.2174/1389557520999201214234735] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 11/22/2022]
Abstract
Quinoline derivatives are considered broad-spectrum pharmacological compounds that exhibit a wide range of biological activities. Integration of quinoline moiety can improve its physical and chemical properties and also pharmacological behavior. Due to its wide range of pharmaceutical applications, it is a very popular compound to design new drugs for the treatment of multiple diseases like cancer, dengue fever, malaria, tuberculosis, fungal infections, AIDS, Alzheimer's disease and diabetes. In this review, our major focus is to pay attention to the biological activities of quinoline compounds in the treatment of these diseases such as anti-viral, anti-cancer, anti-malarial, antibacterial, anti-fungal, anti-tubercular and anti-diabetic.
Collapse
Affiliation(s)
- Rukhsana Tabassum
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, 36100, Pakistan
| | - Muhammad Ashfaq
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, 36100, Pakistan
| | - Hiroyuki Oku
- Division of Molecular Science, Graduate School of Science & Engineering Gunma University, Gunma 376-8515, Japan
| |
Collapse
|
9
|
Yuan J, Liu Z, Zhang Z, Yan D, Zhang W. Synthesis and biological evaluation of naphthoquinone phenacylimidazolium derivatives. Bioorg Med Chem Lett 2021; 41:127977. [PMID: 33766771 DOI: 10.1016/j.bmcl.2021.127977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/14/2021] [Indexed: 01/19/2023]
Abstract
In order to expand structural diversity and improve antitumor efficiency, forty new naphthoquinone phenacylimidazolium derivatives were designed, synthesized and evaluated. Good synthetic yields were obtained under mild conditions using easily available starting materials. Cytotoxicity of these compounds was evaluated in vitro against a panel of human tumor cell lines: human breast carcinoma cell lines (MCF-7), human cervical carcinoma cell lines (HeLa), and human lung carcinoma cell lines (A549). Among them, the optimal compound 7m showed splendid antiproliferative activity with low to 50 nM IC50 values against MCF-7 and excellent selectivity of 256-fold compared with the normal cell lines L929. Compound 7m induced apoptosis in a dose-dependent manner. Further mechanism experiments showed that compound 7m dramatically inhibited the expression of survivin and activated the pro-apoptotic protein caspase-3. Our results indicated that the structural modification on the 1,3-substituents of naphthoquinone imidazoliums without 2-substituent is also promising to obtain new antitumor compounds.
Collapse
Affiliation(s)
- Jing Yuan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Frontier Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Zhanxiong Liu
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Frontier Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Zhenfeng Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| | - Deyue Yan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Frontier Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Wanbin Zhang
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Frontier Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China; School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| |
Collapse
|
10
|
Valderrama JA, Ríos D, Muccioli GG, Buc Calderon P, Benites J. In Vitro Inhibition of Hsp90 Protein by Benzothiazoloquinazolinequinones Is Enhanced in The Presence of Ascorbate. A Preliminary In Vivo Antiproliferative Study. Molecules 2020; 25:E953. [PMID: 32093392 PMCID: PMC7071032 DOI: 10.3390/molecules25040953] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/16/2020] [Accepted: 02/18/2020] [Indexed: 01/14/2023] Open
Abstract
A series of benzo[g]benzothiazolo[2,3-b]quinazoline-7,12-quinones were prepared from 2-acylnaphthohydroquinones and 2-aminobenzothiazoles and were evaluated for their in vitro antiproliferative activity. After screening using the MTT reduction assay, their IC50 values were calculated on a panel of cancer cells (T24, DU-145, MCF-7). Current standard anticancer drugs were included as control, and their calculated IC50 values were 7.8 and 23.5 µM for 5-fluorouracil and tamoxifen, respectively. Non-cancer cells (AG1523) were included to assess cancer cell sensitivity and drug selectivity. Four members of the series, with IC50 values from 0.11 to 2.98 µM, were chosen for further assays. The selected quinones were evaluated regarding their effects on cancer cell proliferation (clonogenic assay) and on Hsp90 and poly(ADPribose)polymerase (PARP) protein integrity. The most active compound (i.e., 15) substantially inhibited colony forming unit (CFU) formation at 0.25 µM. In the presence of ascorbate, it induced an oxidative cleavage of Hsp90 but had no effect on PARP protein integrity. In an in vivo animal model, it discreetly increased the mean survival time (m.s.t.) of tumor-bearing mice. In light of these results, compound 15 represents a potential lead-molecule to be further developed.
Collapse
Affiliation(s)
- Jaime A. Valderrama
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Casilla 121, Iquique 1100000, Chile; (D.R.); (P.B.C.)
- Instituto de Ciencias Exactas y Naturales, Universidad Arturo Prat, Casilla 121, Iquique 1100000, Chile
| | - David Ríos
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Casilla 121, Iquique 1100000, Chile; (D.R.); (P.B.C.)
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids (BPBL), Louvain Drug Research Institute, Université catholique de Louvain, 72 Avenue E. Mounier, BPBL 7201, 1200 Brussels, Belgium;
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Casilla 121, Iquique 1100000, Chile; (D.R.); (P.B.C.)
- Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, 73 Avenue E. Mounier, 1200 Brussels, Belgium
| | - Julio Benites
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Casilla 121, Iquique 1100000, Chile; (D.R.); (P.B.C.)
- Instituto de Ciencias Exactas y Naturales, Universidad Arturo Prat, Casilla 121, Iquique 1100000, Chile
| |
Collapse
|
11
|
Ahenkorah S, Coertzen D, Tong JX, Fridianto K, Wittlin S, Birkholtz LM, Tan KSW, Lam Y, Go ML, Haynes RK. Antimalarial N 1, N 3-Dialkyldioxonaphthoimidazoliums: Synthesis, Biological Activity, and Structure-activity Relationships. ACS Med Chem Lett 2020; 11:49-55. [PMID: 31938463 DOI: 10.1021/acsmedchemlett.9b00457] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/11/2019] [Indexed: 12/19/2022] Open
Abstract
Here we report the nanomolar potencies of N 1,N 3-dialkyldioxonaphthoimidazoliums against asexual forms of sensitive and resistant Plasmodium falciparum. Activity was dependent on the presence of the fused quinone-imidazolium entity and lipophilicity imparted by the N1/N3 alkyl residues on the scaffold. Gametocytocidal activity was also detected, with most members active at IC50 < 1 μM. A representative analog with good solubility, limited PAMPA permeability, and microsomal stability demonstrated oral efficacy on a humanized mouse model of P. falciparum.
Collapse
Affiliation(s)
| | - Dina Coertzen
- Institute for Sustainable Malaria Control, Department of Biochemistry, Genetics and Microbiology, University of Pretoria, 0028 Pretoria, South Africa
| | | | | | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
- University of Basel, 4003 Basel, Switzerland
| | - Lyn-Marie Birkholtz
- Institute for Sustainable Malaria Control, Department of Biochemistry, Genetics and Microbiology, University of Pretoria, 0028 Pretoria, South Africa
| | | | | | | | - Richard K. Haynes
- Centre of Excellence for Pharmaceutical Sciences, North-West University, 2531 Potchefstroom, South Africa
| |
Collapse
|
12
|
Neri JM, Cavalcanti LN, Araújo RM, Menezes FG. 2,3-Dichloroquinoxaline as a versatile building block for heteroaromatic nucleophilic substitution: A review of the last decade. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2017.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
13
|
Choudhari D, Salunke-Gawali S, Chakravarty D, Shaikh SR, Lande DN, Gejji SP, Rao PK, Satpute S, Puranik VG, Gonnade R. Synthesis and biological activity of imidazole based 1,4-naphthoquinones. NEW J CHEM 2020. [DOI: 10.1039/c9nj04339j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Design and development of drugs in multi-drug resistant (MDR) infections have been of growing interest. The syntheses, structural studies, antibacterial and antifungal activities of imidazole-based 1,4-naphthoquinones are studied in this investigation.
Collapse
Affiliation(s)
- Dinkar Choudhari
- Department of Chemistry
- Savitribai Phule Pune University
- Pune 411007
- India
| | | | | | - Samir R. Shaikh
- Centre for Material Characterization
- National Chemical Laboratory
- Pune 411008
- India
| | - Dipali N. Lande
- Department of Chemistry
- Savitribai Phule Pune University
- Pune 411007
- India
| | - Shridhar P. Gejji
- Department of Chemistry
- Savitribai Phule Pune University
- Pune 411007
- India
| | - Pradeep Kumar Rao
- Department of Chemistry
- Savitribai Phule Pune University
- Pune 411007
- India
| | - Surekha Satpute
- Department of Microbiology
- Savitribai Phule Pune University
- Pune 411007
- India
| | - Vedavati G. Puranik
- Centre for Material Characterization
- National Chemical Laboratory
- Pune 411008
- India
| | - Rajesh Gonnade
- Centre for Material Characterization
- National Chemical Laboratory
- Pune 411008
- India
| |
Collapse
|
14
|
Chiou JT, Lee YC, Huang CH, Shi YJ, Wang LJ, Chang LS. Autophagic HuR mRNA degradation induces survivin and MCL1 downregulation in YM155-treated human leukemia cells. Toxicol Appl Pharmacol 2020; 387:114857. [DOI: 10.1016/j.taap.2019.114857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 12/24/2022]
|
15
|
Go YH, Lim C, Jeong HC, Kwon OS, Chung S, Lee H, Kim W, Suh YG, Son WS, Lee MO, Cha HJ, Kim SH. Structure-Activity Relationship Analysis of YM155 for Inducing Selective Cell Death of Human Pluripotent Stem Cells. Front Chem 2019; 7:298. [PMID: 31157201 PMCID: PMC6532689 DOI: 10.3389/fchem.2019.00298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/12/2019] [Indexed: 12/16/2022] Open
Abstract
Despite great potential for regenerative medicine, the high tumorigenic potential of human pluripotent stem cells (hPSCs) to form undesirable teratoma is an important technical hurdle preventing safe cell therapy. Various small molecules that induce the complete elimination of undifferentiated hPSCs, referred to as "stemotoxics," have been developed to facilitate tumor-free cell therapy, including the Survivin inhibitor YM155. In the present work, based on the chemical structure of YM155, total 26 analogs were synthesized and tested for stemotoxic activity toward human embryonic stem cells (hESCs) and induced PSCs (iPSCs). We found that a hydrogen bond acceptor in the pyrazine ring of YM155 derivatives is critical for stemotoxic activity, which is completely lost in hESCs lacking SLC35F2, which encodes a solute carrier protein. These results suggest that hydrogen bonding interactions between the nitrogens of the pyrazine ring and the SLC35F2 protein are critical for entry of YM155 into hPSCs, and hence stemotoxic activity.
Collapse
Affiliation(s)
- Young-Hyun Go
- Department of Life Sciences, College of Natural Sciences, Sogang University, Seoul, South Korea
| | - Changjin Lim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pochen-si, South Korea
| | - Ho-Chang Jeong
- Department of Life Sciences, College of Natural Sciences, Sogang University, Seoul, South Korea
| | - Ok-Seon Kwon
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Sungkyun Chung
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pochen-si, South Korea
| | - Haeseung Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Wankyu Kim
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Young-Ger Suh
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pochen-si, South Korea
| | - Woo Sung Son
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pochen-si, South Korea
| | - Mi-Ok Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Hyuk-Jin Cha
- College of Pharmacy, Seoul National University, Seoul, South Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Seok-Ho Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pochen-si, South Korea
| |
Collapse
|
16
|
Jang HJ, Chung IY, Lim C, Chung S, Kim BO, Kim ES, Kim SH, Cho YH. Redirecting an Anticancer to an Antibacterial Hit Against Methicillin-Resistant Staphylococcus aureus. Front Microbiol 2019; 10:350. [PMID: 30858845 PMCID: PMC6398426 DOI: 10.3389/fmicb.2019.00350] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/11/2019] [Indexed: 01/18/2023] Open
Abstract
YM155 is a clinically evaluated anticancer with a fused naphthoquinone-imidazolium scaffold. In this study, we demonstrated that based on weak or cryptic antibacterial activity of YM155 against methicillin-resistant Staphylococcus aureus (MRSA) (MIC of 50 μg/ml), some congeneric compounds with short alkyl chains (e.g., c5 with a hexyl chain) at the N3 position of the scaffold, displayed more potent antibacterial activity against MRSA (MIC of 3.13 μg/ml), which is in a clinically achievable range. Their antibacterial activity was evident against Gram-negative bacteria, only in the presence of the outer membrane-permeabilizing agent, polymyxin B. The antibacterial efficacy of c5 was confirmed using the Drosophila systemic infection model. We also characterized five spontaneous c5-resistant MRSA mutants that carry mutations in the ubiE gene, for quinone metabolism and respiratory electron transfer, and subsequently exhibited reduced respiration activity. The antibacterial activity of c5 was compromised either by an antioxidant, N-acetylcysteine, or in an anaerobic condition. These suggest that the antibacterial mechanism of c5 involves the generation of reactive oxygen species (ROS), presumably during respiratory electron transport. This study provides an insight into "drug redirecting," through a chemical modification, based on an ROS-generating pharmacophore.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - In-Young Chung
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Changjin Lim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Sungkyun Chung
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Bi-O Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Eun Sook Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Seok-Ho Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - You-Hee Cho
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| |
Collapse
|
17
|
Jafari F, Baghayi H, Lavaee P, Hadizadeh F, Soltani F, Moallemzadeh H, Mirzaei S, Aboutorabzadeh SM, Ghodsi R. Design, synthesis and biological evaluation of novel benzo- and tetrahydrobenzo-[h]quinoline derivatives as potential DNA-intercalating antitumor agents. Eur J Med Chem 2019; 164:292-303. [DOI: 10.1016/j.ejmech.2018.12.060] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 12/08/2018] [Accepted: 12/24/2018] [Indexed: 12/15/2022]
|
18
|
Chen BJ, Mani V, Huang ST, Hu YC, Shan HCP. Bisintercalating DNA redox reporters for real-time electrochemical qLAMP. Biosens Bioelectron 2018; 129:277-283. [PMID: 30266426 DOI: 10.1016/j.bios.2018.09.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/30/2018] [Accepted: 09/16/2018] [Indexed: 11/19/2022]
Abstract
The electrochemical detection methods have emerged as a potential alternative to the bench-top optical systems in monitoring nucleic acid amplification. DNA intercalating redox reporters play a crucial role in such monitoring schemes. Here, a series of bisintercalating redox probes have been tailor-made to meet specific requirements of electrochemical quantitative loop-mediated isothermal amplification (qLAMP). The probes composed of two naphthoquinone-imidazole (NQIM) derivatives as signal motifs that are covalently bridged by different linkers (R). They are bis-NQIM-R; R = Alkane (Ak), ethylene glycol (EG) and phenyl (Ph). The linkers allow the probes to be fine-tuned for securing ideal redox reporter. DNA binding studies via electrochemical and fluorescence techniques demonstrate that the bis-NQIM-R probes possess better ds-DNA bisintercalating ability compared to their mono-analogs. The bis-NQIM-Ph was implemented in a real-time electrochemical qLAMP, for which a prototype custom-made device that can perform fully automated multiplexed analyses is devised. A single copy of Salmonella DNA was quantified in just 10 min and the performance is comparable to the benchtop fluorescence method. Thus, the bisintercalating redox reporters incorporated electrochemical detection schemes hold great promise in qLAMP.
Collapse
Affiliation(s)
- Bo-Jun Chen
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, ROC
| | - Veerappan Mani
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, ROC; Graduate Institute of Biomedical and Biochemical Engineering, National Taipei University of Technology, Taipei, Taiwan, ROC
| | - Sheng-Tung Huang
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan, ROC; Graduate Institute of Biomedical and Biochemical Engineering, National Taipei University of Technology, Taipei, Taiwan, ROC.
| | - Yi-Chiuen Hu
- National Applied Research Lab, Hsinchu, Taiwan, ROC
| | | |
Collapse
|
19
|
Pan L, Zheng Q, Chen Y, Yang R, Yang Y, Li Z, Meng X. Design, synthesis and biological evaluation of novel naphthoquinone derivatives as IDO1 inhibitors. Eur J Med Chem 2018; 157:423-436. [DOI: 10.1016/j.ejmech.2018.08.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/07/2018] [Accepted: 08/04/2018] [Indexed: 11/24/2022]
|
20
|
Wani TH, Surendran S, Jana A, Chakrabarty A, Chowdhury G. Quinone-Based Antitumor Agent Sepantronium Bromide (YM155) Causes Oxygen-Independent Redox-Activated Oxidative DNA Damage. Chem Res Toxicol 2018; 31:612-618. [PMID: 29897742 DOI: 10.1021/acs.chemrestox.8b00094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Sepantronium bromide (YM155) is a small molecule antitumor agent currently in phase II clinical trials. Although developed as survivin suppressor, YM155's primary mode of action has recently been found to be DNA damage. However, the mechanism of DNA damage by YM155 is still unknown. Knowing the mechanism of action of an anticancer drug is necessary to formulate a rational drug combination and select a cancer type for achieving maximum clinical efficacy. Using cell-based assays, we showed that YM155 causes extensive DNA cleavage and reactive oxygen species generation. DNA cleavage by YM155 was found to be inhibited by radical scavengers and desferal. The reducing agent DTT and the cellular reducing system xanthine/xanthine oxidase were found to reductively activate YM155 and cause DNA cleavage. Unlike quinones, DNA cleavage by YM155 occurs in the presence of catalase and under hypoxic conditions, indicating that hydrogen peroxide and oxygen are not necessary. Although YM155 is a quinone, it does not follow a typical quinone mechanism. Consistent with these observations, a mechanism has been proposed that suggests that YM155 can cause oxidative DNA cleavage upon 2-electron reductive activation.
Collapse
|
21
|
Liu Z, Zhang Z, Zhang W, Yan D. 2-Substituted-1-(2-morpholinoethyl)-1H-naphtho[2,3-d]imidazole-4,9-diones: Design, synthesis and antiproliferative activity. Bioorg Med Chem Lett 2018; 28:2454-2458. [PMID: 29884536 DOI: 10.1016/j.bmcl.2018.06.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/30/2018] [Accepted: 06/02/2018] [Indexed: 12/30/2022]
Abstract
Thirty-six of novel compounds 2-substituted-1-(2-morpholinoethyl)-1H-naphtho[2,3-d]imidazole-4,9-diones, bearing a N-(2-morpholinoethyl) group and a 2-substituted imidazole segment on a naphthoquinone skeleton, were designed, synthesized and tested as anticancer agents. Cytotoxicity was evaluated in vitro against three human cancer cell lines: human breast carcinoma cell line (MCF-7), human cervical carcinoma cell line (Hela), and human lung carcinoma cell line (A549); and one normal cell line: mouse fibroblast cell line (L929). Among them, the compound 2-(3-chloro-4-methoxyphenyl)-1-(2-morpholinoethyl)-1H-naphtho[2,3-d]imidazole-4,9-dione showed good antiproliferative activity against MCF-7, Hela and A549 (IC50 values are equal to 10.6 μM, 8.3 μM and 4.3 μM respectively) and low cytotoxicity to L929 (IC50 value is equal to 67.3 μM).
Collapse
Affiliation(s)
- Zhanxiong Liu
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Zhenfeng Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Wanbin Zhang
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China; School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| | - Deyue Yan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| |
Collapse
|
22
|
Theoretical study of the intermolecular recognition mechanism between Survivin and substrate based on conserved binding mode analysis. J Mol Graph Model 2018; 83:53-63. [PMID: 29772403 DOI: 10.1016/j.jmgm.2018.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/02/2018] [Accepted: 05/02/2018] [Indexed: 01/12/2023]
Abstract
Survivin is the smallest member of IAP (inhibitor of apoptosis protein) family, which plays important roles in both mitosis and apoptosis. It has become an attractive drug target due to its overexpression in many human cancers. Survivin has been proven to bind to Smac/DIABLO protein that indirectly inhibits apoptosis. Meanwhile, it is the key subunit of chromosome passenger complex (CPC) which bind to the N-terminal tail of phosphorylated histone H3T3ph during mitosis. Up to now, Survivin directly targeting inhibitor has yet to merge since the difficulty of disrupting the protein-protein interactions (PPIs) between Survivin and its substrate proteins. Nevertheless, currently known binding partners of Survivin provide crucial information about conserved recognition mechanism, which can assist in the detection of some uncharted substrates and also the Survivin inhibitors. Herein, we adopted a method that using four substrates to analyze the common binding mode of Survivin. To accomplish this, conventional molecular dynamics (MD) simulations, molecular mechanics/generalized born surface area (MM-GBSA) binding free energy calculations and energy decomposition were carried out to assess the binding affinity and per-residue contributions. We found that there are two anchor sites of Survivin responsible for maintaining the binding conformation and one sub-pocket for intermolecular recognition. The results of this study synthetically describe the binding mechanism and provide valuable guidance for rational drug design of PPI inhibitor.
Collapse
|
23
|
Dioxonaphthoimidazoliums AB1 and YM155 disrupt phosphorylation of p50 in the NF-κB pathway. Oncotarget 2017; 7:11625-36. [PMID: 26872379 PMCID: PMC4905498 DOI: 10.18632/oncotarget.7299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 01/27/2016] [Indexed: 12/11/2022] Open
Abstract
The NF-κB pathway is overexpressed in non-small cell lung cancers (NSCLC) and contributes to the poor prognosis and high mortality characterizing this malignancy. Silencing the p50 and p65 NF-κB subunits in the NSCLC H1299 cell line led to profound loss in cell viability and downregulated anti-apoptotic proteins survivin and Mcl1. We also showed that a survivin suppressant, the dioxonaphthoimidazolium YM155, and its structural analog AB1 arrested the growth of H1299 cells at nanomolar concentrations. Both compounds were apoptogenic and suppressed survivin and other anti-apoptotic proteins (Mcl1, Bcl-2, Bcl-xl) in a dose- and/or time-dependent manner. YM155 and AB1 did not affect the expression of key proteins (IκBα, p65, p50) involved in NF-κB signaling. Stable IκBα levels suggest that the NF-κB/IκB complex and proteins upstream of IκBα, were not targeted. Neither did the compounds intercept the nuclear translocation of the p50 and p65 subunits. On the other hand, YM155 and AB1 suppressed the phosphorylation of the p50 subunit at Ser337 which is critical in promoting the binding of NF-κB dimers to DNA. Both compounds duly impeded the binding of NF-κB dimers to DNA and attenuated transcriptional activity of luciferase-transfected HEK293 cells controlled by NF-κB response elements. We propose that the “silencing” the NF-κB pathway effected by these compounds contributed to their potent apoptogenic effects on H1299. Notwithstanding, the mechanism(s) involved in their ability to abolish phosphorylation of p50 remains to be elucidated. Taken together, these results disclose a novel facet of functionalized dioxonaphthoimidazoliums that could account for their potent cell killing property.
Collapse
|
24
|
Nyquist MD, Corella A, Burns J, Coleman I, Gao S, Tharakan R, Riggan L, Cai C, Corey E, Nelson PS, Mostaghel EA. Exploiting AR-Regulated Drug Transport to Induce Sensitivity to the Survivin Inhibitor YM155. Mol Cancer Res 2017; 15:521-531. [PMID: 28465296 PMCID: PMC5471626 DOI: 10.1158/1541-7786.mcr-16-0315-t] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/03/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022]
Abstract
Androgen receptor (AR) signaling is fundamental to prostate cancer and is the dominant therapeutic target in metastatic disease. However, stringent androgen deprivation therapy regimens decrease quality of life and have been largely unsuccessful in curtailing mortality. Recent clinical and preclinical studies have taken advantage of the dichotomous ability of AR signaling to elicit growth-suppressive and differentiating effects by administering hyperphysiologic levels of testosterone. In this study, high-throughput drug screening identified a potent synergy between high-androgen therapy and YM155, a transcriptional inhibitor of survivin (BIRC5). This interaction was mediated by the direct transcriptional upregulation of the YM155 transporter SLC35F2 by the AR. Androgen-mediated YM155-induced cell death was completely blocked by the overexpression of multidrug resistance transporter ABCB1. SLC35F2 expression was significantly correlated with intratumor androgen levels in four distinct patient-derived xenograft models, and with AR activity score in a large gene expression dataset of castration-resistant metastases. A subset of tumors had significantly elevated SLC35F2 expression and, therefore, may identify patients who are highly responsive to YM155 treatment. IMPLICATIONS The combination of androgen therapy with YM155 represents a novel drug synergy, and SLC35F2 may serve as a clinical biomarker of response to YM155.
Collapse
Affiliation(s)
- Michael D Nyquist
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Alexandra Corella
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - John Burns
- Virginia Mason Medical Center, Seattle, Washington
| | - Ilsa Coleman
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Shuai Gao
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Robin Tharakan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Luke Riggan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Changmeng Cai
- Center for Personalized Cancer Therapy, University of Massachusetts Boston, Boston, Massachusetts
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Urology, University of Washington, Seattle, Washington
- Division of Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Elahe A Mostaghel
- Division of Oncology, Department of Medicine, University of Washington, Seattle, Washington.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
25
|
Nyquist MD, Prasad B, Mostaghel EA. Harnessing Solute Carrier Transporters for Precision Oncology. Molecules 2017; 22:E539. [PMID: 28350329 PMCID: PMC5570559 DOI: 10.3390/molecules22040539] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Solute Carrier (SLC) transporters are a large superfamily of transmembrane carriers involved in the regulated transport of metabolites, nutrients, ions and drugs across cellular membranes. A subset of these solute carriers play a significant role in the cellular uptake of many cancer therapeutics, ranging from chemotherapeutics such as antimetabolites, topoisomerase inhibitors, platinum-based drugs and taxanes to targeted therapies such as tyrosine kinase inhibitors. SLC transporters are co-expressed in groups and patterns across normal tissues, suggesting they may comprise a coordinated regulatory circuit serving to mediate normal tissue functions. In cancer however, there are dramatic changes in expression patterns of SLC transporters. This frequently serves to feed the increased metabolic demands of the tumor cell for amino acids, nucleotides and other metabolites, but also presents a therapeutic opportunity, as increased transporter expression may serve to increase intracellular concentrations of substrate drugs. In this review, we examine the regulation of drug transporters in cancer and how this impacts therapy response, and discuss novel approaches to targeting therapies to specific cancers via tumor-specific aberrations in transporter expression. We propose that among the oncogenic changes in SLC transporter expression there exist emergent vulnerabilities that can be exploited therapeutically, extending the application of precision medicine from tumor-specific drug targets to tumor-specific determinants of drug uptake.
Collapse
Affiliation(s)
- Michael D Nyquist
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA.
| | - Elahe A Mostaghel
- Division of Oncology, Department of Medicine, University of Washington, Seattle, WA 98195 USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
26
|
Novel multi-substituted benzyl acridone derivatives as survivin inhibitors for hepatocellular carcinoma treatment. Eur J Med Chem 2017; 129:337-348. [PMID: 28237663 DOI: 10.1016/j.ejmech.2017.02.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/07/2017] [Accepted: 02/10/2017] [Indexed: 11/24/2022]
Abstract
Sorafenib was the only small-molecule drug approved by FDA for treatment of the advanced hepatocellular carcinoma (HCC). Recent study indicated that YM155 was a promising agent for HCC cells with high survivin expression, however, the antitumor activity needs to be further improved. Based on molecular docking and rational design method, a series of multi-substituted benzyl acridone derivatives were designed and synthesized. MTT assay indicated that some of the synthesized compounds displayed better antiproliferative activity against HepG2 cells than YM155. Later study indicated that the representive compound 8u may directly interact with survivin protein and induce HepG2 cells apoptosis, which is different from YM155. In addition, ADME property was predicted in silico, and it performed well. Moreover, in vivo preliminary experiments showed that 8u may be a good lead compound in the treatment of HCC.
Collapse
|
27
|
Ho SHS, Ali A, Ng YC, Lam KKM, Wang S, Chan WK, Chin TM, Go ML. Dioxonaphthoimidazoliums are Potent and Selective Rogue Stem Cell Clearing Agents with SOX2-Suppressing Properties. ChemMedChem 2016; 11:1944-55. [PMID: 27444266 DOI: 10.1002/cmdc.201600262] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 06/27/2016] [Indexed: 01/11/2023]
Abstract
Pluripotent stem cells are uniquely positioned for regenerative medicine, but their clinical potential can only be realized if their tumorigenic tendencies are decoupled from their pluripotent properties. Deploying small molecules to remove remnant undifferentiated pluripotent cells, which would otherwise transform into teratomas and teratomacarcinomas, offers several advantages over non-pharmacological methods. Dioxonapthoimidazolium YM155, a survivin suppressant, induced selective and potent cell death of undifferentiated stem cells. Herein, the structural requirements for stemotoxicity were investigated and found to be closely aligned with those essential for cytotoxicity in malignant cells. There was a critical reliance on the quinone and imidazolium moieties but a lesser dependence on ring substituents, which served mainly to fine-tune activity. Several potent analogues were identified which, like YM155, suppressed survivin and decreased SOX2 in stem cells. The decrease in SOX2 would cause an imbalance in pluripotent factors that could potentially prompt cells to differentiate and hence decrease the risk of aberrant teratoma formation. As phosphorylation of the NF-κB p50 subunit was also suppressed, the crosstalk between phospho-p50, SOX2, and survivin could implicate a causal role for NF-κB signaling in mediating the stem cell clearing properties of dioxonaphthoimidazoliums.
Collapse
Affiliation(s)
- Si-Han Sherman Ho
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Azhar Ali
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
| | - Yi-Cheng Ng
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Kuen-Kuen Millie Lam
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Shu Wang
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.,Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore, 138669, Singapore
| | - Woon-Khiong Chan
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Tan-Min Chin
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
| | - Mei-Lin Go
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore.
| |
Collapse
|