1
|
Li Y, Liu H, Fang R, Jin J, Yang F, Chen J, Zhang J. Designing novel Au(III) complexes based on the structure of diazepam: Achieving a multiaction mechanism against glioma. Eur J Med Chem 2025; 283:117171. [PMID: 39705733 DOI: 10.1016/j.ejmech.2024.117171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/01/2024] [Accepted: 12/12/2024] [Indexed: 12/22/2024]
Abstract
Metal-based drugs have been used in the clinical treatment of tumors for over 30 years. However, no metal-based drugs have been clinically approved to treat glioma. Although metal complexes have excellent cytotoxicity, their most critical problem is crossing the blood-brain barrier. Therefore, to enable metal complexes to cross blood-brain barrier and target glioma therapy, herein, we propose to rationally used the basic structure of diazepam (5-chlorobenzophenone) and thiosemicarbazide to synthesize gold (Au) complexes C1, C2 and C3 with antiglioma activity. The C3 complex with two methyl groups attached to the N3 of thiosemicarbazone exhibited excellent cytotoxicity to glioma cells through its multiaction mechanism against glioma, inducing apoptosis, autophagy death, and deoxyribonucleic acid damage. In addition, the synthesized C3 complex can effectively cross the blood-brain barrier and accumulate in glioma, considerably decreasing the untoward reaction in vivo. Our findings provide a novel strategy for designing metal-based complexes for the treatment of glioma.
Collapse
Affiliation(s)
- Yanping Li
- Mental Health Education Center of College Student, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Haoran Liu
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Ronghao Fang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, 541004, PR China
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| | - Juzheng Zhang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| |
Collapse
|
2
|
Cui Q, Ding W, Luo B, Lu W, Huang P, Wen S. Novel gold-based complex GC7 suppresses cancer cell proliferation via impacting energy metabolism mediated by mitochondria. Bioorg Med Chem 2024; 112:117897. [PMID: 39216383 DOI: 10.1016/j.bmc.2024.117897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Due to their pivotal roles in regulating energy metabolism and apoptosis, mitochondria in cancer cells have been considered a vulnerable and feasible target. Many anticancer agents, e.g., metal-based compounds, are found to target and disturb mitochondria primarily, which may lead to the disturbance of energy metabolism and, more importantly, the initiation of apoptosis. In this work, a gold-based complex 7 (GC7) was synthesized and evaluated in a series of different cancer cell lines. The anticancer efficacies of GC7 on cell viability, apoptosis, and colony formation were determined. Cellular thioredoxin reductase (TrxR) activity, oxygen consumption rate (OCR), glucose uptake, and lactate production following GC7 treatment were evaluated and analyzed. The Jeko-1 and A549 xenograft models were used to assess GC7's tumor-suppressing effects. The results showed that GC7 possessed a broad-spectrum anticancer effect, with IC50 values ranging from 0.43 to 1.2 μM in multiple cancer cell lines, which was more potent than gold-based auranofin (∼2-6 folds). GC7 (0.3 and 1 μM) efficiently induced apoptosis of Jeko-1, A549, and HCT116 cells, and it suppressed the sphere formation of cancer stem cells GSC11 and GSC23 cells at 0.1 μM, and it completely eliminated colony at 0.3 μM. The preliminary mechanistic study showed that GC7 inhibited cellular TrxR activity, suppressed mitochondrial OCR, reduced mitochondrial membrane potential (MMP), decreased glucose uptake, and possibly suppressed glycolysis to reduce lactate production. GC7 was predicted to have a similar yet slightly different pharmacokinetic profile as auranofin. Finally, GC7 (20 mg/kg, oral, 5/week, or 3 mg/kg, IP, 3/week) significantly inhibited tumor growth. In conclusion, GC7 showed great potential in suppressing cancer cell proliferation, probably via inhibiting TrxR and impacting mitochondria-mediated energy metabolism.
Collapse
Affiliation(s)
- Qingbin Cui
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510006, China
| | - Wenwen Ding
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510006, China
| | - Bingling Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510006, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510006, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510006, China.
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510006, China.
| |
Collapse
|
3
|
Zhao Q, Han B, Peng C, Zhang N, Huang W, He G, Li JL. A promising future of metal-N-heterocyclic carbene complexes in medicinal chemistry: The emerging bioorganometallic antitumor agents. Med Res Rev 2024; 44:2194-2235. [PMID: 38591229 DOI: 10.1002/med.22039] [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: 01/19/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/10/2024]
Abstract
Metal complexes based on N-heterocyclic carbene (NHC) ligands have emerged as promising broad-spectrum antitumor agents in bioorganometallic medicinal chemistry. In recent decades, studies on cytotoxic metal-NHC complexes have yielded numerous compounds exhibiting superior cytotoxicity compared to cisplatin. Although the molecular mechanisms of these anticancer complexes are not fully understood, some potential targets and modes of action have been identified. However, a comprehensive review of their biological mechanisms is currently absent. In general, apoptosis caused by metal-NHCs is common in tumor cells. They can cause a series of changes after entering cells, such as mitochondrial membrane potential (MMP) variation, reactive oxygen species (ROS) generation, cytochrome c (cyt c) release, endoplasmic reticulum (ER) stress, lysosome damage, and caspase activation, ultimately leading to apoptosis. Therefore, a detailed understanding of the influence of metal-NHCs on cancer cell apoptosis is crucial. In this review, we provide a comprehensive summary of recent advances in metal-NHC complexes that trigger apoptotic cell death via different apoptosis-related targets or signaling pathways, including B-cell lymphoma 2 (Bcl-2 family), p53, cyt c, ER stress, lysosome damage, thioredoxin reductase (TrxR) inhibition, and so forth. We also discuss the challenges, limitations, and future directions of metal-NHC complexes to elucidate their emerging application in medicinal chemistry.
Collapse
Affiliation(s)
- Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Dermatology & Venerolog, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gu He
- Department of Dermatology & Venerolog, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jun-Long Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Anti-Infective Agent Creation Engineering Research Centre of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, China
| |
Collapse
|
4
|
Wang WJ, Ling YY, Shi Y, Wu XW, Su X, Li ZQ, Mao ZW, Tan CP. Identification of mitochondrial ATP synthase as the cellular target of Ru-polypyridyl- β-carboline complexes by affinity-based protein profiling. Natl Sci Rev 2024; 11:nwae234. [PMID: 39114378 PMCID: PMC11304990 DOI: 10.1093/nsr/nwae234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
Ruthenium polypyridyl complexes are promising anticancer candidates, while their cellular targets have rarely been identified, which limits their clinical application. Herein, we design a series of Ru(II) polypyridyl complexes containing bioactive β-carboline derivatives as ligands for anticancer evaluation, among which Ru5 shows suitable lipophilicity, high aqueous solubility, relatively high anticancer activity and cancer cell selectivity. The subsequent utilization of a photo-clickable probe, Ru5a, serves to validate the significance of ATP synthase as a crucial target for Ru5 through photoaffinity-based protein profiling. Ru5 accumulates in mitochondria, impairs mitochondrial functions and induces mitophagy and ferroptosis. Combined analysis of mitochondrial proteomics and RNA-sequencing shows that Ru5 significantly downregulates the expression of the chloride channel protein, and influences genes related to ferroptosis and epithelial-to-mesenchymal transition. Finally, we prove that Ru5 exhibits higher anticancer efficacy than cisplatin in vivo. We firstly identify the molecular targets of ruthenium polypyridyl complexes using a photo-click proteomic method coupled with a multiomics approach, which provides an innovative strategy to elucidate the anticancer mechanisms of metallo-anticancer candidates.
Collapse
Affiliation(s)
- Wen-Jin Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yu-Yi Ling
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yin Shi
- School of Pharmacy, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Xiao-Wen Wu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xuxian Su
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zheng-Qiu Li
- School of Pharmacy, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, Sun Yat-Sen University, Guangzhou 510006, China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, China
- Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
5
|
Ruan ML, Ni WX, Chu JCH, Lam TL, Law KC, Zhang Y, Yang G, He Y, Zhang C, Fung YME, Liu T, Huang T, Lok CN, Chan SLF, Che CM. Iridium(III) carbene complexes as potent girdin inhibitors against metastatic cancers. Proc Natl Acad Sci U S A 2024; 121:e2316615121. [PMID: 38861602 PMCID: PMC11194514 DOI: 10.1073/pnas.2316615121] [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: 10/06/2023] [Accepted: 04/27/2024] [Indexed: 06/13/2024] Open
Abstract
Many cancer-driving protein targets remain undruggable due to a lack of binding molecular scaffolds. In this regard, octahedral metal complexes with unique and versatile three-dimensional structures have rarely been explored as inhibitors of undruggable protein targets. Here, we describe antitumor iridium(III) pyridinium-N-heterocyclic carbene complex 1a, which profoundly reduces the viability of lung and breast cancer cells as well as cancer patient-derived organoids at low micromolar concentrations. Compound 1a effectively inhibits the growth of non-small-cell lung cancer and triple-negative breast cancer xenograft tumors, impedes the metastatic spread of breast cancer cells, and can be modified into an antibody-drug conjugate payload to achieve precise tumor delivery in mice. Identified by thermal proteome profiling, an important molecular target of 1a in cellulo is Girdin, a multifunctional adaptor protein that is overexpressed in cancer cells and unequivocally serves as a signaling hub for multiple pivotal oncogenic pathways. However, specific small-molecule inhibitors of Girdin have not yet been developed. Notably, 1a exhibits high binding affinity to Girdin with a Kd of 1.3 μM and targets the Girdin-linked EGFR/AKT/mTOR/STAT3 cancer-driving pathway, inhibiting cancer cell proliferation and metastatic activity. Our study reveals a potent Girdin-targeting anticancer compound and demonstrates that octahedral metal complexes constitute an untapped library of small-molecule inhibitors that can fit into the ligand-binding pockets of key oncoproteins.
Collapse
Affiliation(s)
- Mei-Ling Ruan
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan430079, China
| | - Wen-Xiu Ni
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Jacky C. H. Chu
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Tsz-Lung Lam
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kwok-Chung Law
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yiwei Zhang
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Guanya Yang
- AI And Life Sciences Institute (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Ying He
- AI And Life Sciences Institute (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Chunlei Zhang
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yi Man Eva Fung
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Tao Liu
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Tao Huang
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Chun-Nam Lok
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Sharon Lai-Fung Chan
- Department of Applied Biology and Chemical Biology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Chi-Ming Che
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
6
|
Chen Y, Wang C, Qi M, Wei Y, Jiang H, Du Z. Molecular targets of cisplatin in HeLa cells explored through competitive activity-based protein profiling strategy. J Inorg Biochem 2024; 254:112518. [PMID: 38460483 DOI: 10.1016/j.jinorgbio.2024.112518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/23/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Cisplatin is widely used as anticancer drugs, and DNA is considered as the main target. Considering its high affinity towards cysteines and the important role of cystine containing proteins, we applied a competitive activity-based protein profiling strategy to identify protein cysteines that bind with cisplatin in HeLa cells. Living cells were treated with cisplatin at cytotoxic concentrations, then the protein was extracted. After labeling with desthiobiotin iodoacetamide (DBIA) probe, protein was precipitated, digested and isotopically labeled, subsequently the peptides were combined, and the biotinylated cysteine-containing peptides were enriched and quantified by LC-MS/MS. A total of 3571 peptides which originated from 1871 proteins were identified using the DBIA probe. Among them, 46 proteins were screened as targets, including proteins that have been identified as binding proteins by previous study. A novel cisplatin target, calpain-1 (CAPN1), was identified and validated as binding with cisplatin in vitro.
Collapse
Affiliation(s)
- Yi Chen
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chenxi Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meiling Qi
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinyu Wei
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongliang Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhifeng Du
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
7
|
Ramos R, Karaiskou A, Botuha C, Amhaz S, Trichet M, Dingli F, Forté J, Lam F, Canette A, Chaumeton C, Salome M, Chenuel T, Bergonzi C, Meyer P, Bohic S, Loew D, Salmain M, Sobczak-Thépot J. Identification of Cellular Protein Targets of a Half-Sandwich Iridium(III) Complex Reveals Its Dual Mechanism of Action via Both Electrophilic and Oxidative Stresses. J Med Chem 2024; 67:6189-6206. [PMID: 38577779 DOI: 10.1021/acs.jmedchem.3c02000] [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: 04/06/2024]
Abstract
Identification of intracellular targets of anticancer drug candidates provides key information on their mechanism of action. Exploiting the ability of the anticancer (C∧N)-chelated half-sandwich iridium(III) complexes to covalently bind proteins, click chemistry with a bioorthogonal azido probe was used to localize a phenyloxazoline-chelated iridium complex within cells and profile its interactome at the proteome-wide scale. Proteins involved in protein folding and actin cytoskeleton regulation were identified as high-affinity targets. Upon iridium complex treatment, the folding activity of Heat Shock Protein HSP90 was inhibited in vitro and major cytoskeleton disorganization was observed. A wide array of imaging and biochemical methods validated selected targets and provided a multiscale overview of the effects of this complex on live human cells. We demonstrate that it behaves as a dual agent, inducing both electrophilic and oxidative stresses in cells that account for its cytotoxicity. The proposed methodological workflow can open innovative avenues in metallodrug discovery.
Collapse
Affiliation(s)
- Robin Ramos
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, 184 rue du Faubourg Saint Antoine, F-75012 Paris, France
| | - Anthi Karaiskou
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, 184 rue du Faubourg Saint Antoine, F-75012 Paris, France
| | - Candice Botuha
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, 4 place Jussieu, F-75005 Paris, France
| | - Sadek Amhaz
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, 184 rue du Faubourg Saint Antoine, F-75012 Paris, France
| | - Michaël Trichet
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Service d'imagerie cellulaire, F-75005 Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, CurieCoreTech Mass Spectrometry Proteomics, F-75248 Paris, France
| | - Jérémy Forté
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, 4 place Jussieu, F-75005 Paris, France
| | - France Lam
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Service d'imagerie cellulaire, F-75005 Paris, France
| | - Alexis Canette
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Service d'imagerie cellulaire, F-75005 Paris, France
| | - Chloé Chaumeton
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Service d'imagerie cellulaire, F-75005 Paris, France
| | - Murielle Salome
- ESRF, The European Synchrotron Research Facility, F-38043 Grenoble cedex 9, France
| | - Thomas Chenuel
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005 Paris, France
| | - Céline Bergonzi
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005 Paris, France
| | - Philippe Meyer
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005 Paris, France
| | - Sylvain Bohic
- Université Grenoble Alpes, INSERM, UA7 STROBE, Synchrotron Radiation for Biomedicine, F-38400 Saint Martin d'Hères, France
| | - Damarys Loew
- Institut Curie, PSL Research University, CurieCoreTech Mass Spectrometry Proteomics, F-75248 Paris, France
| | - Michèle Salmain
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, 4 place Jussieu, F-75005 Paris, France
| | - Joëlle Sobczak-Thépot
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, 184 rue du Faubourg Saint Antoine, F-75012 Paris, France
| |
Collapse
|
8
|
Coverdale JPC, Polepalli S, Arruda MAZ, da Silva ABS, Stewart AJ, Blindauer CA. Recent Advances in Metalloproteomics. Biomolecules 2024; 14:104. [PMID: 38254704 PMCID: PMC10813065 DOI: 10.3390/biom14010104] [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: 10/20/2023] [Revised: 11/17/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Interactions between proteins and metal ions and their complexes are important in many areas of the life sciences, including physiology, medicine, and toxicology. Despite the involvement of essential elements in all major processes necessary for sustaining life, metalloproteomes remain ill-defined. This is not only owing to the complexity of metalloproteomes, but also to the non-covalent character of the complexes that most essential metals form, which complicates analysis. Similar issues may also be encountered for some toxic metals. The review discusses recently developed approaches and current challenges for the study of interactions involving entire (sub-)proteomes with such labile metal ions. In the second part, transition metals from the fourth and fifth periods are examined, most of which are xenobiotic and also tend to form more stable and/or inert complexes. A large research area in this respect concerns metallodrug-protein interactions. Particular attention is paid to separation approaches, as these need to be adapted to the reactivity of the metal under consideration.
Collapse
Affiliation(s)
- James P. C. Coverdale
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Edgbaston B15 2TT, UK;
| | | | - Marco A. Z. Arruda
- Institute of Chemistry, Department of Analytical Chemistry, Universidade Estadual de Campinas, Campinas 13083-970, Brazil; (M.A.Z.A.); (A.B.S.d.S.)
| | - Ana B. Santos da Silva
- Institute of Chemistry, Department of Analytical Chemistry, Universidade Estadual de Campinas, Campinas 13083-970, Brazil; (M.A.Z.A.); (A.B.S.d.S.)
| | - Alan J. Stewart
- School of Medicine, University of St. Andrews, St Andrews KY16 9TF, UK
| | | |
Collapse
|
9
|
Tabana Y, Babu D, Fahlman R, Siraki AG, Barakat K. Target identification of small molecules: an overview of the current applications in drug discovery. BMC Biotechnol 2023; 23:44. [PMID: 37817108 PMCID: PMC10566111 DOI: 10.1186/s12896-023-00815-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
Target identification is an essential part of the drug discovery and development process, and its efficacy plays a crucial role in the success of any given therapy. Although protein target identification research can be challenging, two main approaches can help researchers make significant discoveries: affinity-based pull-down and label-free methods. Affinity-based pull-down methods use small molecules conjugated with tags to selectively isolate target proteins, while label-free methods utilize small molecules in their natural state to identify targets. Target identification strategy selection is essential to the success of any drug discovery process and must be carefully considered when determining how to best pursue a specific project. This paper provides an overview of the current target identification approaches in drug discovery related to experimental biological assays, focusing primarily on affinity-based pull-down and label-free approaches, and discusses their main limitations and advantages.
Collapse
Affiliation(s)
- Yasser Tabana
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Dinesh Babu
- Li Ka Shing Applied Virology Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Richard Fahlman
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Arno G Siraki
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
10
|
Arojojoye AS, Walker B, Dewahare JC, Afrifa MAO, Parkin S, Awuah SG. Circumventing Physicochemical Barriers of Cyclometalated Gold(III) Dithiocarbamate Complexes with Protein-Based Nanoparticle Delivery to Enhance Anticancer Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43607-43620. [PMID: 37698293 PMCID: PMC11264193 DOI: 10.1021/acsami.3c10025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Optimizing the bioavailability of drug candidates is crucial to successful drug development campaigns, especially for metal-derived chemotherapeutic agents. Nanoparticle delivery strategies can be deployed to overcome physicochemical limitations associated with drugs to improve bioavailability, pharmacokinetics, efficacy, and minimize toxicity. Biodegradable albumin nanoconstructs offer pragmatic solutions for drug delivery of metallodrugs with translational benefits in the clinic. In this work, we explored a logical approach to investigate and resolve the physicochemical drawbacks of gold(III) complexes with albumin nanoparticle delivery to improve solubility, enhance intracellular accumulation, circumvent premature deactivation, and enhance anticancer activity. We synthesized and characterized stable gold(III) dithiocarbamate complexes with a variable degree of cyclometalation such as phenylpyridine (C^N) or biphenyl (C^C) Au(III) framework and different alkyl chain lengths. We noted that extended alkyl chain lengths impaired the solubility of these complexes in biological media, thus adversely impacting potency. Encapsulation of these complexes in bovine serum albumin (BSA) reversed solubility limitations and improved cancer cytotoxicity by ∼25-fold. Further speciation and mechanism of action studies demonstrate the stability of the compounds and alteration of mitochondria bioenergetics, respectively. We postulate that this nanodelivery strategy is a relevant approach for translational small-molecule gold drug delivery.
Collapse
Affiliation(s)
| | - Breyanna Walker
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | - James C. Dewahare
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | | | - Sean Parkin
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA.
- Center for Pharmaceutical Research and Innovation and Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington KY 40536
| |
Collapse
|
11
|
Borutzki Y, Skos L, Gerner C, Meier‐Menches SM. Exploring the Potential of Metal-Based Candidate Drugs as Modulators of the Cytoskeleton. Chembiochem 2023; 24:e202300178. [PMID: 37345897 PMCID: PMC10946712 DOI: 10.1002/cbic.202300178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/23/2023]
Abstract
During recent years, accumulating evidence suggested that metal-based candidate drugs are promising modulators of cytoskeletal and cytoskeleton-associated proteins. This was substantiated by the identification and validation of actin, vimentin and plectin as targets of distinct ruthenium(II)- and platinum(II)-based modulators. Despite this, structural information about molecular interaction is scarcely available. Here, we compile the scattered reports about metal-based candidate molecules that influence the cytoskeleton, its associated proteins and explore their potential to interfere in cancer-related processes, including proliferation, invasion and the epithelial-to-mesenchymal transition. Advances in this field depend crucially on determining binding sites and on gaining comprehensive insight into molecular drug-target interactions. These are key steps towards establishing yet elusive structure-activity relationships.
Collapse
Affiliation(s)
- Yasmin Borutzki
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Doctoral School of ChemistryUniversity of Vienna1090ViennaAustria
| | - Lukas Skos
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Doctoral School of ChemistryUniversity of Vienna1090ViennaAustria
| | - Christopher Gerner
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Samuel M. Meier‐Menches
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| |
Collapse
|
12
|
Bourehil L, Soep C, Seng S, Dutrannoy S, Igoudjil S, Forté J, Gontard G, Lesage D, Bertrand B, Dossmann H. Bond-Dissociation Energies to Probe Pyridine Electronic Effects on Organogold(III) Complexes: From Methodological Developments to Application in π-Backdonation Investigation and Catalysis. Inorg Chem 2023; 62:13304-13314. [PMID: 37560906 DOI: 10.1021/acs.inorgchem.3c01584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
In this work, we report on the synthesis of several organogold(III) complexes based on 4,4'-diterbutylbiphenyl (C^C) and 2,6-bis(4-terbutylphenyl)pyridine (C^N^C) ligands and bond with variously substituted pyridine ligands (pyrR). Altogether, 33 complexes have been prepared and studied with mass spectrometry using higher-energy collision dissociation (HCD) in an Orbitrap mass spectrometer. A complete methodology including the kinetic modeling of the dissociation process based on the Rice-Ramsperger-Kassel-Marcus (RRKM) statistical method is proposed to obtain critical energies E0 of the pyrR loss for all complexes. The capacity of these E0 values to describe the pyridine ligand effect is further explored, at the same time as more classical descriptors such as 1H pyridinic NMR shift variation upon coordination and Au-NpyrR bond length measured by X-ray diffraction. An extensive theoretical work, including density functional theory (DFT) and domain-based local pair natural orbital coupled-cluster theory (DLPNO-CCSD(T)) methods, is also carried out to provide bond-dissociation energies, which are compared to experimental results. Results show that dissociation energy outperforms other descriptors, in particular to describe ligand effects over a large electronic effect range as seen by confronting the results to the pyrR pKa values. Further insights into the Au-NpyrR bond are obtained through an energy decomposition analysis (EDA) study, which confirms the isolobal character of Au+ with H+. Finally, the correlation between the lability of the pyridine ligands toward the catalytic efficiency of the complexes could be demonstrated in an intramolecular hydroarylation reaction of alkyne. The results were rationalized considering both pre-catalyst activation and catalyst reactivity. This study establishes the possibility of correlating dissociation energy, which is a gas-phase descriptor, with condensed-phase parameters such as catalysis efficiency. It therefore holds great potential for inorganic and organometallic chemistry by opening a convenient and easy way to evaluate the electronic influence of a ligand toward a metallic center.
Collapse
Affiliation(s)
- Lyna Bourehil
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
- Synchrotron SOLEIL, L'Orme des Merisiers, St Aubin, BP 48, F-91192 Gif-sur-Yvette, France
| | - Clément Soep
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
| | - Sopheak Seng
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
- Institute of Physical Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 2, D-76131 Karlsruhe, Germany
| | - Sarah Dutrannoy
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
| | - Stacy Igoudjil
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
| | - Jérémy Forté
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
| | - Geoffrey Gontard
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
| | - Denis Lesage
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
| | - Benoît Bertrand
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
| | - Héloïse Dossmann
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, IPCM, F-75005 Paris, France
| |
Collapse
|
13
|
Gukathasan S, Obisesan OA, Saryazdi S, Ratliff L, Parkin S, Grossman RB, Awuah SG. A Conformationally Restricted Gold(III) Complex Elicits Antiproliferative Activity in Cancer Cells. Inorg Chem 2023; 62:13118-13129. [PMID: 37530672 PMCID: PMC11268950 DOI: 10.1021/acs.inorgchem.3c02066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Diamine ligands are effective structural scaffolds for tuning the reactivity of transition-metal complexes for catalytic, materials, and phosphorescent applications and have been leveraged for biological use. In this work, we report the synthesis and characterization of a novel class of cyclometalated [C^N] Au(III) complexes bearing secondary diamines including a norbornane backbone, (2R,3S)-N2,N3-dibenzylbicyclo[2.2.1]heptane-2,3-diamine, or a cyclohexane backbone, (1R,2R)-N1,N2-dibenzylcyclohexane-1,2-diamine. X-ray crystallography confirms the square-planar geometry and chirality at nitrogen. The electronic character of the conformationally restricted norbornane backbone influences the electrochemical behavior with redox potentials of -0.8 to -1.1 V, atypical for Au(III) complexes. These compounds demonstrate promising anticancer activity, particularly, complex 1, which bears a benzylpyridine organogold framework, and supported by the bicyclic conformationally restricted diaminonorbornane, shows good potency in A2780 cells. We further show that a cellular response to 1 evokes reactive oxygen species (ROS) production and does not induce mitochondrial dysfunction. This class of complexes provides significant stability and reactivity for different applications in protein modification, catalysis, and therapeutics.
Collapse
Affiliation(s)
| | | | - Setareh Saryazdi
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
| | - Libby Ratliff
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
| | - Sean Parkin
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
| | - Robert B. Grossman
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
| | - Samuel G. Awuah
- Department of Chemistry, University of Kentucky, Lexington KY 40506, USA
- Center for Pharmaceutical Research and Innovation and Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington KY 40536, USA
- Markey Cancer Center, University of Kentucky, Lexington KY 40536
| |
Collapse
|
14
|
Geri A, Massai L, Messori L. Protein Metalation by Medicinal Gold Compounds: Identification of the Main Features of the Metalation Process through ESI MS Experiments. Molecules 2023; 28:5196. [PMID: 37446857 DOI: 10.3390/molecules28135196] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Gold compounds form a new class of promising anticancer agents with innovative modes of action. It is generally believed that anticancer gold compounds, at variance with clinically established platinum drugs, preferentially target proteins rather than nucleic acids. The reactions of several gold compounds with a few model proteins have been systematically explored in recent years through ESI MS measurements to reveal adduct formation and identify the main features of those reactions. Here, we focus our attention on a group of five gold compounds of remarkable medicinal interest, i.e., Auranofin, Au(NHC)Cl, [Au(NHC)2]PF6, Aubipyc, and Auoxo6, and on their reactions with four different biomolecular targets, i.e., the proteins HEWL, hCA I, HSA and the C-terminal dodecapeptide of the enzyme thioredoxin reductase. Complete ESI MS data are available for those reactions due to previous experimental work conducted in our laboratory. From the comparative analysis of the ESI MS reaction profiles, some characteristic trends in the metallodrug-protein reactivity may be identified as detailed below. The main features are described and analyzed in this review. Overall, all these observations are broadly consistent with the concept that cytotoxic gold drugs preferentially target cancer cell proteins, with a remarkable selectivity for the cysteine and selenocysteine proteome. These interactions typically result in severe damage to cancer cell metabolism and profound alterations in the redox state, leading to eventual cancer cell death.
Collapse
Affiliation(s)
- Andrea Geri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Florence, Italy
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Florence, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Florence, Italy
| |
Collapse
|
15
|
Li SR, Tan YM, Zhang L, Zhou CH. Comprehensive Insights into Medicinal Research on Imidazole-Based Supramolecular Complexes. Pharmaceutics 2023; 15:1348. [PMID: 37242590 PMCID: PMC10222694 DOI: 10.3390/pharmaceutics15051348] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The electron-rich five-membered aromatic aza-heterocyclic imidazole, which contains two nitrogen atoms, is an important functional fragment widely present in a large number of biomolecules and medicinal drugs; its unique structure is beneficial to easily bind with various inorganic or organic ions and molecules through noncovalent interactions to form a variety of supramolecular complexes with broad medicinal potential, which is being paid an increasing amount of attention regarding more and more contributions to imidazole-based supramolecular complexes for possible medicinal application. This work gives systematical and comprehensive insights into medicinal research on imidazole-based supramolecular complexes, including anticancer, antibacterial, antifungal, antiparasitic, antidiabetic, antihypertensive, and anti-inflammatory aspects as well as ion receptors, imaging agents, and pathologic probes. The new trend of the foreseeable research in the near future toward imidazole-based supramolecular medicinal chemistry is also prospected. It is hoped that this work provides beneficial help for the rational design of imidazole-based drug molecules and supramolecular medicinal agents and more effective diagnostic agents and pathological probes.
Collapse
Affiliation(s)
- Shu-Rui Li
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Yi-Min Tan
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ling Zhang
- School of Chemical Technology, Shijiazhuang University, Shijiazhuang 050035, China
| | - Cheng-He Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| |
Collapse
|
16
|
Skos L, Borutzki Y, Gerner C, Meier-Menches SM. Methods to identify protein targets of metal-based drugs. Curr Opin Chem Biol 2023; 73:102257. [PMID: 36599256 DOI: 10.1016/j.cbpa.2022.102257] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 01/03/2023]
Abstract
Metal-based anticancer agents occupy a distinct chemical space due to their particular coordination geometry and reactivity. Despite the initial DNA-targeting paradigm for this class of compounds, it is now clear that they can also be tuned to target proteins in cells, depending on the metal and ligand scaffold. Since metallodrug discovery is dominated by phenotypic screenings, tailored proteomics strategies were crucial to identify and validate protein targets of several investigative and clinically advanced metal-based drugs. Here, such experimental approaches are discussed, which showed that metallodrugs based on ruthenium, gold, rhenium and even platinum, can selectively and specifically target proteins with clear-cut down-stream effects. Target identification strategies are expected to support significantly the mechanism-driven clinical translation of metal-based drugs.
Collapse
Affiliation(s)
- Lukas Skos
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Doctoral School of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Yasmin Borutzki
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Doctoral School of Chemistry, University of Vienna, 1090 Vienna, Austria; Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Joint Metabolome Facility, University of Vienna and Medical University Vienna, 1090 Vienna, Austria
| | - Samuel M Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Joint Metabolome Facility, University of Vienna and Medical University Vienna, 1090 Vienna, Austria.
| |
Collapse
|
17
|
Moreno-Alcántar G, Picchetti P, Casini A. Gold Complexes in Anticancer Therapy: From New Design Principles to Particle-Based Delivery Systems. Angew Chem Int Ed Engl 2023; 62:e202218000. [PMID: 36847211 DOI: 10.1002/anie.202218000] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 02/28/2023]
Abstract
The discovery of the medicinal properties of gold complexes has fuelled the design and synthesis of new anticancer metallodrugs, which have received special attention due to their unique modes of action. Current research in the development of gold compounds with therapeutic properties is predominantly focused on the molecular design of drug leads with superior pharmacological activities, e.g., by introducing targeting features. Moreover, intensive research aims at improving the physicochemical properties of gold compounds, such as chemical stability and solubility in the physiological environment. In this regard, the encapsulation of gold compounds in nanocarriers or their chemical grafting onto targeted delivery vectors could lead to new nanomedicines that eventually reach clinical applications. Herein, we provide an overview of the state-of-the-art progress of gold anticancer compounds, andmore importantly we thoroughly revise the development of nanoparticle-based delivery systems for gold chemotherapeutics.
Collapse
Affiliation(s)
- Guillermo Moreno-Alcántar
- Chair of Medicinal and Bioinorganic Chemistry, School of Natural Sciences, Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching b. München, Germany
| | - Pierre Picchetti
- Karlsruhe Institute of Technology (KIT), Institute of Nanotechnology, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, School of Natural Sciences, Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching b. München, Germany
| |
Collapse
|
18
|
Ma Z, Han H, Zhao Y. Mitochondrial dysfunction-targeted nanosystems for precise tumor therapeutics. Biomaterials 2023; 293:121947. [PMID: 36512861 DOI: 10.1016/j.biomaterials.2022.121947] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Mitochondria play critical roles in the regulation of the proliferation and apoptosis of cancerous cells. Targeted induction of mitochondrial dysfunction in cancer cells by multifunctional nanosystems for cancer treatment has attracted increasing attention in the past few years. Numerous therapeutic nanosystems have been designed for precise tumor therapy by inducing mitochondrial dysfunction, including reducing adenosine triphosphate, breaking redox homeostasis, inhibiting glycolysis, regulating proteins, membrane potential depolarization, mtDNA damage, mitophagy dysregulation and so on. Understanding the mechanisms of mitochondrial dysfunction would be helpful for efficient treatment of diseases and accelerating the translation of these therapeutic strategies into the clinic. Then, various strategies to construct mitochondria-targeted nanosystems and induce mitochondrial dysfunction are summarized, and the recent research progress regarding precise tumor therapeutics is highlighted. Finally, the major challenges and an outlook in this rapidly developing field are discussed. This review is expected to inspire further development of novel mitochondrial dysfunction-based strategies for precise treatments of cancer and other human diseases.
Collapse
Affiliation(s)
- Zhaoyu Ma
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, PR China; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Heyou Han
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, PR China.
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore.
| |
Collapse
|
19
|
Abstract
Environmental agents of exposure can damage proteins, affecting protein function and cellular protein homeostasis. Specific residues are inherently chemically susceptible to damage from individual types of exposure. Amino acid content is not completely predictive of protein susceptibility, as secondary, tertiary, and quaternary structures of proteins strongly influence the reactivity of the proteome to individual exposures. Because we cannot readily predict which proteins will be affected by which chemical exposures, mass spectrometry-based proteomic strategies are necessary to determine the protein targets of environmental toxins and toxicants. This review describes the mechanisms by which environmental exposure to toxins and toxicants can damage proteins and affect their function, and emerging omic methodologies that can be used to identify the protein targets of a given agent. These methods include target identification strategies that have recently revolutionized the drug discovery field, such as activity-based protein profiling, protein footprinting, and protein stability profiling technologies. In particular, we highlight the necessity of multiple, complementary approaches to fully interrogate how protein integrity is challenged by individual exposures.
Collapse
Affiliation(s)
- Joseph C Genereux
- Department of Chemistry, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
20
|
Ikram M, Rehman S, Feroz I, Farzia, Khan R, Sinnokrot MO, Subhan F, Naeem M, Schulzke C. Synthesis, spectral, Hirshfeld surface analysis and biological evaluation of a Schiff base copper(II) complex: towards a copper(II) based human anti-glioblastoma agent. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.134960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
21
|
Yan X, Zhou Y, Li H, Jiang G, Sun H. Metallomics and metalloproteomics. COMPREHENSIVE INORGANIC CHEMISTRY III 2023:53-76. [DOI: 10.1016/b978-0-12-823144-9.00060-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
22
|
De Franco M, Saab M, Porchia M, Marzano C, Nolan SP, Nahra F, Van Hecke K, Gandin V. Unveiling the Potential of Innovative Gold(I) and Silver(I) Selenourea Complexes as Anticancer Agents Targeting TrxR and Cellular Redox Homeostasis. Chemistry 2022; 28:e202201898. [PMID: 36106679 PMCID: PMC10092581 DOI: 10.1002/chem.202201898] [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: 06/20/2022] [Indexed: 11/11/2022]
Abstract
A series of NHC-based selenourea Ag(I) and Au(I) complexes were evaluated for their anticancer potential in vitro, on 2D and 3D human cancer cell systems. All NHC-based selenourea complexes possess an outstanding cytotoxic potency, which was comparable or even better than that of the reference metallodrug auranofin, and were also able to overcome both platinum-based and multi-drug resistances. Intriguingly, their cytotoxic potency did not correlate with solution stability, partition coefficient or cellular uptake. On the other hand, mechanistic studies in cancer cells revealed their ability to strongly and selectively inhibit the redox-regulating enzyme Thioredoxin Reductase (TrxR), being even more effective than auranofin, a well-known TrxR inhibitor, without affecting other redox enzymes such as Glutathione Reductase (GR). The inhibition of TrxR in H157 human cancer cells caused, in turn, the disruption of cellular thiol-redox homeostasis and of mitochondria pathophysiology, ultimately leading to cancer cell death through apoptosis.
Collapse
Affiliation(s)
- Michele De Franco
- Dipartimento di Scienze del FarmacoUniversità degli Studi di PadovaVia F. Marzolo 5I-35131PadovaItaly
| | - Marina Saab
- Department of ChemistryCenter for Sustainable Chemistry Ghent UniversityKrigsman 281, Building S39000 GhentBelgium
| | | | - Cristina Marzano
- Dipartimento di Scienze del FarmacoUniversità degli Studi di PadovaVia F. Marzolo 5I-35131PadovaItaly
| | - Steven P. Nolan
- Department of ChemistryCenter for Sustainable Chemistry Ghent UniversityKrigsman 281, Building S39000 GhentBelgium
| | - Fady Nahra
- Department of ChemistryCenter for Sustainable Chemistry Ghent UniversityKrigsman 281, Building S39000 GhentBelgium
- VITO (Flemish Institute for Technological Research)Boeretang 2002400MolBelgium
| | - Kristof Van Hecke
- Department of ChemistryCenter for Sustainable Chemistry Ghent UniversityKrigsman 281, Building S39000 GhentBelgium
| | - Valentina Gandin
- Dipartimento di Scienze del FarmacoUniversità degli Studi di PadovaVia F. Marzolo 5I-35131PadovaItaly
| |
Collapse
|
23
|
Neuditschko B, King AP, Huang Z, Janker L, Bileck A, Borutzki Y, Marker SC, Gerner C, Wilson JJ, Meier‐Menches SM. An Anticancer Rhenium Tricarbonyl Targets Fe-S Cluster Biogenesis in Ovarian Cancer Cells. Angew Chem Int Ed Engl 2022; 61:e202209136. [PMID: 36004624 PMCID: PMC9827826 DOI: 10.1002/anie.202209136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Indexed: 01/12/2023]
Abstract
Target identification remains a critical challenge in inorganic drug discovery to deconvolute potential polypharmacology. Herein, we describe an improved approach to prioritize candidate protein targets based on a combination of dose-dependent chemoproteomics and treatment effects in living cancer cells for the rhenium tricarbonyl compound TRIP. Chemoproteomics revealed 89 distinct dose-dependent targets with concentrations of competitive saturation between 0.1 and 32 μM despite the broad proteotoxic effects of TRIP. Target-response networks revealed two highly probable targets of which the Fe-S cluster biogenesis factor NUBP2 was competitively saturated by free TRIP at nanomolar concentrations. Importantly, TRIP treatment led to a down-regulation of Fe-S cluster containing proteins and upregulated ferritin. Fe-S cluster depletion was further verified by assessing mitochondrial bioenergetics. Consequently, TRIP emerges as a first-in-class modulator of the scaffold protein NUBP2, which disturbs Fe-S cluster biogenesis at sub-cytotoxic concentrations in ovarian cancer cells.
Collapse
Affiliation(s)
- Benjamin Neuditschko
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Present address: Institute Krems BioanalyticsIMC University of Applied Sciences Krems3500KremsAustria
| | - A. Paden King
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY 14853USA,Present address: Chemical Biology LaboratoryCenter for Cancer ResearchNational Cancer InstituteFrederickMD 21702USA
| | - Zhouyang Huang
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY 14853USA
| | - Lukas Janker
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Andrea Bileck
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Yasmin Borutzki
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
| | - Sierra C. Marker
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY 14853USA,Present address: Chemical Biology LaboratoryCenter for Cancer ResearchNational Cancer InstituteFrederickMD 21702USA
| | - Christopher Gerner
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Justin J. Wilson
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY 14853USA
| | - Samuel M. Meier‐Menches
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| |
Collapse
|
24
|
Zhang J, Li Y, Fang R, Wei W, Wang Y, Jin J, Yang F, Chen J. Organometallic gold(I) and gold(III) complexes for lung cancer treatment. Front Pharmacol 2022; 13:979951. [PMID: 36176441 PMCID: PMC9513137 DOI: 10.3389/fphar.2022.979951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Metal compounds, especially gold complexes, have recently gained increasing attention as possible lung cancer therapeutics. Some gold complexes display not only excellent activity in cisplatin-sensitive lung cancer but also in cisplatin-resistant lung cancer, revealing promising prospects in the development of novel treatments for lung cancer. This review summarizes examples of anticancer gold(I) and gold (III) complexes for lung cancer treatment, including mechanisms of action and approaches adopted to improve their efficiency. Several excellent examples of gold complexes against lung cancer are highlighted.
Collapse
Affiliation(s)
- Juzheng Zhang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Yanping Li
- School of Public Health, Guilin Medical University, Guilin, China
| | - Ronghao Fang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Wei Wei
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Yong Wang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, China
- *Correspondence: Feng Yang, mailto:, Jian Chen, mailto:
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
- *Correspondence: Feng Yang, mailto:, Jian Chen, mailto:
| |
Collapse
|
25
|
Neuditschko B, King AP, Huang Z, Janker L, Bileck A, Borutzki Y, Marker SC, Gerner C, Wilson JJ, Meier-Menches SM. An Anticancer Rhenium Tricarbonyl Targets Fe‐S Cluster Biogenesis in Ovarian Cancer Cells. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Benjamin Neuditschko
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - A. Paden King
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Zhouyang Huang
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Lukas Janker
- University of Vienna Faculty of Chemistry: Universitat Wien Fakultat fur Chemie Department of Analytical Chemistry AUSTRIA
| | - Andrea Bileck
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - Yasmin Borutzki
- University of Vienna: Universitat Wien Institute of Inorganic Chemistry AUSTRIA
| | - Sierra C. Marker
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Christopher Gerner
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - Justin J. Wilson
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Samuel M. Meier-Menches
- University of Vienna: Universitat Wien Department of Analytical Chemistry Waehringer Str. 38 1090 Vienna AUSTRIA
| |
Collapse
|
26
|
Jiang M, Zhang Z, Li W, Man X, Sun H, Liang H, Yang F. Developing a Copper(II) Agent Based on His-146 and His-242 Residues of Human Serum Albumin Nanoparticles: Integration To Overcome Cisplatin Resistance and Inhibit the Metastasis of Nonsmall Cell Lung Cancer. J Med Chem 2022; 65:9447-9458. [PMID: 35786921 DOI: 10.1021/acs.jmedchem.2c00698] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To overcome the resistance of nonsmall cell lung cancer (NSCLC) cells to cisplatin and inhibit their metastasis, we proposed to develop a Cu(II) agent based on the specific residue(s) of HSA nanoparticles (NPs) for multitargeting the tumor microenvironment (TME). To this end, we not only synthesized four Cu(II) 2-hydroxy-3-methoxybenzaldehyde thiosemicarbazone compounds (C1-C4), obtaining a Cu compound (C4) with significant cytotoxicity by studying their structure-activity relationships, but also revealed the binding mechanism of C4 to HSA through X-ray crystallography and confirmed the successful construction of a new HSA-C4 NPs delivery system. C4 and HSA-C4 NPs inhibited the A549cisR tumor growth and metastasis, and HSA NPs optimized the anticancer behavior of C4. We further confirmed the anticancer mechanism of the C4/HSA-C4 NP multitargeting TME to overcome cisplatin resistance: killing tumor cells by acting on the mtDNA and inducing apoptosis, polarizing M2-type macrophages to the M1-type, and inhibiting angiogenesis.
Collapse
Affiliation(s)
- Ming Jiang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China.,School of Food and Biochemical Engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Zhenlei Zhang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Wenjuan Li
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Xueyu Man
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Hong Liang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Feng Yang
- School of Chemistry and Pharmaceutical Sciences, State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| |
Collapse
|
27
|
Broad Spectrum Functional Activity of Structurally Related Monoanionic Au(III) Bis(Dithiolene) Complexes. Int J Mol Sci 2022; 23:ijms23137146. [PMID: 35806151 PMCID: PMC9266914 DOI: 10.3390/ijms23137146] [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: 05/23/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 01/27/2023] Open
Abstract
The biological properties of sixteen structurally related monoanionic gold (III) bis(dithiolene/diselenolene) complexes were evaluated. The complexes differ in the nature of the heteroatom connected to the gold atom (AuS for dithiolene, AuSe for diselenolene), the substituent on the nitrogen atom of the thiazoline ring (Me, Et, Pr, iPr and Bu), the nature of the exocyclic atom or group of atoms (O, S, Se, C(CN)2) and the counter-ion (Ph4P+ or Et4N+). The anticancer and antimicrobial activities of all the complexes were investigated, while the anti-HIV activity was evaluated only for selected complexes. Most complexes showed relevant anticancer activities against Cisplatin-sensitive and Cisplatin-resistant ovarian cancer cells A2780 and OVCAR8, respectively. After 48 h of incubation, the IC50 values ranged from 0.1–8 µM (A2780) and 0.8–29 µM (OVCAR8). The complexes with the Ph4P+ ([P]) counter-ion are in general more active than their Et4N+ ([N]) analogues, presenting IC50 values in the same order of magnitude or even lower than Auranofin. Studies in the zebrafish embryo model further showed that, despite their marked anticancer effect, the complexes with [P] counter-ion exhibited low in vivo toxicity. In general, the exocyclic exchange of sulfur by oxygen or ylidenemalononitrile (C(CN)2) enhanced the compounds toxicity. Most complexes containing the [P] counter ion exhibited exceptional antiplasmodial activity against the Plasmodium berghei parasite liver stages, with submicromolar IC50 values ranging from 400–700 nM. In contrast, antibacterial/fungi activities were highest for most complexes with the [N] counter-ion. Auranofin and two selected complexes [P][AuSBu(=S)] and [P][AuSEt(=S)] did not present anti-HIV activity in TZM-bl cells. Mechanistic studies for selected complexes support the idea that thioredoxin reductase, but not DNA, is a possible target for some of these complexes. The complexes [P] [AuSBu(=S)], [P] [AuSEt(=S)], [P] [AuSEt(=Se)] and [P] [AuSeiPr(=S)] displayed a strong quenching of the fluorescence intensity of human serum albumin (HSA), which indicates a strong interaction with this protein. Overall, the results highlight the promising biological activities of these complexes, warranting their further evaluation as future drug candidates with clinical applicability.
Collapse
|
28
|
Guadagno L, Raimondo M, Vertuccio L, Lamparelli EP, Ciardulli MC, Longo P, Mariconda A, Della Porta G, Longo R. Electrospun Membranes Designed for Burst Release of New Gold-Complexes Inducing Apoptosis of Melanoma Cells. Int J Mol Sci 2022; 23:ijms23137147. [PMID: 35806152 PMCID: PMC9267035 DOI: 10.3390/ijms23137147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 01/27/2023] Open
Abstract
Two non-commercial metallic Au-based complexes were tested against one of the most aggressive malignant melanomas of the skin (MeWo cells), through cell viability and time-lapse live-cell imaging system assays. The tests with the complexes were carried out both in the form of free metallic complexes, directly in contact with the MeWo cell line culture, and embedded in fibers of Polycaprolactone (PCL) membranes produced by the electrospinning technique. Membranes functionalized with complexes were prepared to evaluate the efficiency of the membranes against the melanoma cells and therefore their feasibility in the application as an antitumoral patch for topical use. Both series of tests highlighted a very effective antitumoral activity, manifesting a very relevant cell viability inhibition after both 24 h and 48 h. In the case of the AuM1 complex at the concentration of 20 mM, melanoma cells completely died in this short period of time. A mortality of around 70% was detected from the tests performed using the membranes functionalized with AuM1 complex at a very low concentration (3 wt.%), even after 24 h of the contact period. The synthesized complexes also manifest high selectivity with respect to the MeWo cells. The peculiar structural and morphological organization of the nanofibers constituting the membranes allows for a very effective antitumoral activity in the first 3 h of treatment. Experimental points of the release profiles were perfectly fitted with theoretical curves, which easily allow interpretation of the kinetic phenomena occurring in the release of the synthesized complexes in the chosen medium.
Collapse
Affiliation(s)
- Liberata Guadagno
- Department of Industrial Engineering, University of Salerno, 84084 Fisciano, Italy;
- Correspondence: (L.G.); (R.L.)
| | - Marialuigia Raimondo
- Department of Industrial Engineering, University of Salerno, 84084 Fisciano, Italy;
| | - Luigi Vertuccio
- Department of Engineering, University of Campania “Luigi Vanvitelli”, 813031 Aversa, Italy;
| | - Erwin Pavel Lamparelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy; (E.P.L.); (M.C.C.); (G.D.P.)
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy; (E.P.L.); (M.C.C.); (G.D.P.)
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, 84084 Fisciano, Italy;
| | | | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy; (E.P.L.); (M.C.C.); (G.D.P.)
- Interdepartment Centre BIONAM, Università di Salerno, 84084 Fisciano, Italy
| | - Raffaele Longo
- Department of Industrial Engineering, University of Salerno, 84084 Fisciano, Italy;
- Correspondence: (L.G.); (R.L.)
| |
Collapse
|
29
|
Tialiou A, Chin J, Keppler BK, Reithofer MR. Current Developments of N-Heterocyclic Carbene Au(I)/Au(III) Complexes toward Cancer Treatment. Biomedicines 2022; 10:biomedicines10061417. [PMID: 35740438 PMCID: PMC9219884 DOI: 10.3390/biomedicines10061417] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022] Open
Abstract
Since their first discovery, N-heterocyclic carbenes have had a significant impact on organometallic chemistry. Due to their nature as strong σ-donor and π-acceptor ligands, they are exceptionally well suited to stabilize Au(I) and Au(III) complexes in biological environments. Over the last decade, the development of rationally designed NHCAu(I/III) complexes to specifically target DNA has led to a new “gold rush” in bioinorganic chemistry. This review aims to summarize the latest advances of NHCAu(I/III) complexes that are able to interact with DNA. Furthermore, the latest advancements on acyclic diamino carbene gold complexes with anticancer activity are presented as these typically overlooked NHC alternatives offer great additional design possibilities in the toolbox of carbene-stabilized gold complexes for targeted therapy.
Collapse
Affiliation(s)
- Alexia Tialiou
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Jiamin Chin
- Institute of Inorganic Chemistry—Functional Materials, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
- Correspondence: (J.C.); (M.R.R.)
| | - Bernhard K. Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Michael R. Reithofer
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Correspondence: (J.C.); (M.R.R.)
| |
Collapse
|
30
|
Lu Y, Ma X, Chang X, Liang Z, Lv L, Shan M, Lu Q, Wen Z, Gust R, Liu W. Recent development of gold(I) and gold(III) complexes as therapeutic agents for cancer diseases. Chem Soc Rev 2022; 51:5518-5556. [PMID: 35699475 DOI: 10.1039/d1cs00933h] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Metal complexes have demonstrated significant antitumor activities and platinum complexes are well established in the clinical application of cancer chemotherapy. However, the platinum-based treatment of different types of cancers is massively hampered by severe side effects and resistance development. Consequently, the development of novel metal-based drugs with different mechanism of action and pharmaceutical profile attracts modern medicinal chemists to design and synthesize novel metal-based agents. Among non-platinum anticancer drugs, gold complexes have gained considerable attention due to their significant antiproliferative potency and efficacy. In most situations, the gold complexes exhibit anticancer activities by targeting thioredoxin reductase (TrxR) or other thiol-rich proteins and enzymes and trigger cell death via reactive oxygen species (ROS). Interestingly, gold complexes were recently reported to elicit biochemical hallmarks of immunogenic cell death (ICD) as an ICD inducer. In this review, the recent progress of gold(I) and gold(III) complexes is comprehensively summarized, and their activities and mechanism of action are documented.
Collapse
Affiliation(s)
- Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaoyan Ma
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xingyu Chang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Zhenlin Liang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Lin Lv
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Min Shan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Qiuyue Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Zhenfan Wen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ronald Gust
- Institute of Pharmacy/Pharmaceutical Chemistry, University of Innsbruck, Center for Chemistry and Biomedicine, Innsbruck, Austria.
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China. .,State key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
31
|
Xiong X, Huang KB, Wang Y, Cao B, Luo Y, Chen H, Yang Y, Long Y, Liu M, Chan ASC, Liang H, Zou T. Target Profiling of an Iridium(III)-Based Immunogenic Cell Death Inducer Unveils the Engagement of Unfolded Protein Response Regulator BiP. J Am Chem Soc 2022; 144:10407-10416. [PMID: 35658433 DOI: 10.1021/jacs.2c02435] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clinical chemotherapeutic drugs have occasionally been observed to induce antitumor immune responses beyond the direct cytotoxicity. Such effects are coined as immunogenic cell death (ICD), representing a "second hit" from the host immune system to tumor cells. Although chemo-immunotherapy is highly promising, ICD inducers remain sparse with vague drug-target mechanisms. Here, we report an endoplasmic reticulum stress-inducing cyclometalated Ir(III)-bisNHC complex (1a) as a new ICD inducer, and based on this compound, a clickable photoaffinity probe was designed for target identification, which unveiled the engagement of the master regulator protein BiP (binding immunoglobulin protein)/GRP78 of the unfolded protein response pathway. This has been confirmed by a series of cellular and biochemical studies including fluorescence microscopy, cellular thermal shift assay, enzymatic assays, and so forth, showing the capability of 1a for BiP destabilization. Notably, besides 1a, the previously reported ICD inducers including KP1339, mitoxantrone, and oxaliplatin were also found to engage BiP interaction, suggesting the important role of BiP in eliciting anticancer immunity. We believe that the ICD-related target information in this work will help to understand the mode of action of ICD that is beneficial to designing new ICD agents with high specificity and improved efficacy.
Collapse
Affiliation(s)
- Xiaolin Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Ke-Bin Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmacy, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Yuan Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Bei Cao
- Warshel Institute for Computational Biology, and General Education Division, The Chinese University of Hong Kong, Shenzhen 518172, P. R. China
| | - Yunli Luo
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Huowen Chen
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Yang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Long
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Moyi Liu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Albert S C Chan
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry & Pharmacy, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Taotao Zou
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
32
|
Jiang J, Cao B, Chen Y, Luo H, Xue J, Xiong X, Zou T. Alkylgold(III) Complexes Undergo Unprecedented Photo-Induced β-Hydride Elimination and Reduction for Targeted Cancer Therapy. Angew Chem Int Ed Engl 2022; 61:e202201103. [PMID: 35165986 DOI: 10.1002/anie.202201103] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Indexed: 11/07/2022]
Abstract
Spatiotemporally controllable activation of prodrugs within tumors is highly desirable for cancer therapy to minimize toxic side effects. Herein we report that stable alkylgold(III) complexes can undergo unprecedented photo-induced β-hydride elimination, releasing alkyl ligands and forming gold(III)-hydride intermediates that could be quickly converted into bioactive [AuIII -S] adducts; meanwhile, the remaining alkylgold(III) complexes can photo-catalytically reduce [AuIII -S] into more bioactive AuI species. Such photo-reactivities make it possible to functionalize gold complexes on the auxiliary alkyl ligands without attenuating the metal-biomacromolecule interactions. As a result, the gold(III) complexes containing glucose-functionalized alkyl ligands displayed efficient and tumor-selective uptake; notably, after one- or two-photon activation, the complexes exhibited high thioredoxin reductase (TrxR) inhibition, potent cytotoxicity, and strong antiangiogenesis and antitumor activities in vivo.
Collapse
Affiliation(s)
- Jia Jiang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Bei Cao
- Warshel Institute for Computational Biology, and General Education Division, The Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
| | - Yuting Chen
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Hejiang Luo
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Jiaying Xue
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Xiaolin Xiong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Taotao Zou
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| |
Collapse
|
33
|
Hylland KT, Schmidtke IL, Wragg DS, Nova A, Tilset M. Synthesis of substituted (N,C) and (N,C,C) Au(III) complexes: the influence of sterics and electronics on cyclometalation reactions. Dalton Trans 2022; 51:5082-5097. [PMID: 35262546 DOI: 10.1039/d2dt00371f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cyclometalated Au(III) complexes are of interest due to their catalytic, medicinal, and photophysical properties. Herein, we describe the synthesis of derivatives of the type (N,C)Au(OAcF)2 (OAcF = trifluoroacetate) and (N,C,C)AuOAcF by a cyclometalation route, where (N,C) and (N,C,C) are chelating 2-arylpyridine ligands. The scope of the synthesis is explored by substituting the 2-arylpyridine core with electron donor or acceptor substituents at one or both rings. Notably, a variety of functionalized Au(III) complexes can be obtained in one step from the corresponding ligand and Au(OAc)3, eliminating the need for organomercury intermediates, which is commonly reported for similar syntheses. The influence of substituents in the ligand backbone on the resulting complexes was assessed using DFT calculations, 15N NMR spectroscopy and single-crystal X-ray diffraction analysis. A correlation between the electronic properties of the (N,C) ligands and their ability to undergo cyclometalation was found from experimental studies combined with natural charge analysis, suggesting the cyclometalation at Au(III) to take place via an electrophilic aromatic substitution-type mechanism. The formation of Au(III) pincer complexes from tridentate (N,C,C) ligands was investigated by synthesis and DFT calculations, in order to assess the feasibility of C(sp3)-H bond activation as a synthetic pathway to (N,C,C) cyclometalated Au(III) complexes. It was found that C(sp3)-H bond activation is feasible for ligands containing different alkyl groups (isopropyl and ethyl), although the C-H activation is less energetically favored compared to a ligand containing tert-butyl groups.
Collapse
Affiliation(s)
- Knut T Hylland
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, N-0315 Oslo, Norway. .,Centre for Materials Science and Nanotechnology, University of Oslo, P.O. Box 1126 Blindern, N-0316 Oslo, Norway
| | - Inga L Schmidtke
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, N-0315 Oslo, Norway. .,Centre for Materials Science and Nanotechnology, University of Oslo, P.O. Box 1126 Blindern, N-0316 Oslo, Norway
| | - David S Wragg
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, N-0315 Oslo, Norway. .,Centre for Materials Science and Nanotechnology, University of Oslo, P.O. Box 1126 Blindern, N-0316 Oslo, Norway
| | - Ainara Nova
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, N-0315 Oslo, Norway. .,Centre for Materials Science and Nanotechnology, University of Oslo, P.O. Box 1126 Blindern, N-0316 Oslo, Norway.,Hylleraas Centre for Quantum Molecular Sciences, Department of Chemistry, University of Oslo, N-0315 Oslo, Norway.,UiT-The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Mats Tilset
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, N-0315 Oslo, Norway. .,Centre for Materials Science and Nanotechnology, University of Oslo, P.O. Box 1126 Blindern, N-0316 Oslo, Norway.,Hylleraas Centre for Quantum Molecular Sciences, Department of Chemistry, University of Oslo, N-0315 Oslo, Norway
| |
Collapse
|
34
|
|
35
|
Gurba A, Taciak P, Sacharczuk M, Młynarczuk-Biały I, Bujalska-Zadrożny M, Fichna J. Gold (III) Derivatives in Colon Cancer Treatment. Int J Mol Sci 2022; 23:724. [PMID: 35054907 PMCID: PMC8775370 DOI: 10.3390/ijms23020724] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality worldwide. Colorectal cancer (CRC) is the third most frequently diagnosed cancer in men and the second in women. Standard patterns of antitumor therapy, including cisplatin, are ineffective due to their lack of specificity for tumor cells, development of drug resistance, and severe side effects. For this reason, new methods and strategies for CRC treatment are urgently needed. Current research includes novel platinum (Pt)- and other metal-based drugs such as gold (Au), silver (Ag), iridium (Ir), or ruthenium (Ru). Au(III) compounds are promising drug candidates for CRC treatment due to their structural similarity to Pt(II). Their advantage is their relatively good solubility in water, but their disadvantage is an unsatisfactory stability under physiological conditions. Due to these limitations, work is still underway to improve the formula of Au(III) complexes by combining with various types of ligands capable of stabilizing the Au(III) cation and preventing its reduction under physiological conditions. This review summarizes the achievements in the field of stable Au(III) complexes with potential cytotoxic activity restricted to cancer cells. Moreover, it has been shown that not nucleic acids but various protein structures such as thioredoxin reductase (TrxR) mediate the antitumor effects of Au derivatives. The state of the art of the in vivo studies so far conducted is also described.
Collapse
Affiliation(s)
- Agata Gurba
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (P.T.); (M.S.); (M.B.-Z.)
| | - Przemysław Taciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (P.T.); (M.S.); (M.B.-Z.)
| | - Mariusz Sacharczuk
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (P.T.); (M.S.); (M.B.-Z.)
- Department of Genomics, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Izabela Młynarczuk-Biały
- Department for Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland;
| | - Magdalena Bujalska-Zadrożny
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (P.T.); (M.S.); (M.B.-Z.)
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215 Lodz, Poland;
| |
Collapse
|
36
|
Xiang Y, Huang H, Wang D, Du J, Wu D, Xiong W, Hong Y, Chen J, Liao X. Organometallic Au(III) Based Electrochemical Sensor with Wide Anodic Potential Window for Sensitive and Selective Detection of Ochratoxin A. ELECTROANAL 2021. [DOI: 10.1002/elan.202100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yuan Xiang
- Collaborative Innovation Center of Postharvest Key Technology and Quality Safety of Fruits and Vegetables in Jiangxi Province Nanchang 330045 P. R. China
- Research Center of Mycotoxin Jiangxi Agricultural University Nanchang 330045 P. R. China
| | - Hao Huang
- Collaborative Innovation Center of Postharvest Key Technology and Quality Safety of Fruits and Vegetables in Jiangxi Province Nanchang 330045 P. R. China
- Research Center of Mycotoxin Jiangxi Agricultural University Nanchang 330045 P. R. China
| | - Dan Wang
- College of Food Science and Engineering Jiangxi Agricultural University Nanchang 330045 P. R. China
| | - Juan Du
- College of Food Science and Engineering Jiangxi Agricultural University Nanchang 330045 P. R. China
| | - Dongping Wu
- Research Center of Mycotoxin Jiangxi Agricultural University Nanchang 330045 P. R. China
| | - Wanming Xiong
- Research Center of Mycotoxin Jiangxi Agricultural University Nanchang 330045 P. R. China
| | - Yanping Hong
- College of Food Science and Engineering Jiangxi Agricultural University Nanchang 330045 P. R. China
| | - Jinyin Chen
- Collaborative Innovation Center of Postharvest Key Technology and Quality Safety of Fruits and Vegetables in Jiangxi Province Nanchang 330045 P. R. China
- College of Materials Chemistry and Chemical Engineering Pingxiang University Pingxiang 337055 P. R. China
| | - Xiaoning Liao
- Collaborative Innovation Center of Postharvest Key Technology and Quality Safety of Fruits and Vegetables in Jiangxi Province Nanchang 330045 P. R. China
- Research Center of Mycotoxin Jiangxi Agricultural University Nanchang 330045 P. R. China
| |
Collapse
|
37
|
|
38
|
van der Westhuizen D, Bezuidenhout DI, Munro OQ. Cancer molecular biology and strategies for the design of cytotoxic gold(I) and gold(III) complexes: a tutorial review. Dalton Trans 2021; 50:17413-17437. [PMID: 34693422 DOI: 10.1039/d1dt02783b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This tutorial review highlights key principles underpinning the design of selected metallodrugs to target specific biological macromolecules (DNA and proteins). The review commences with a descriptive overview of the eukaryotic cell cycle and the molecular biology of cancer, particularly apoptosis, which is provided as a necessary foundation for the discovery, design, and targeting of metal-based anticancer agents. Drugs which target DNA have been highlighted and clinically approved metallodrugs discussed. A brief history of the development of mainly gold-based metallodrugs is presented prior to addressing ligand systems for stabilizing and adding functionality to bio-active gold(I) and gold(III) complexes, particularly in the burgeoning field of anticancer metallodrugs. Concepts such as multi-modal and selective cytotoxic agents are covered where necessary for selected compounds. The emerging role of carbenes as the ligand system of choice to achieve these goals for gold-based metallodrug candidates is highlighted prior to closing the review with comments on some future directions that this research field might follow. The latter section ultimately emphasizes the importance of understanding the fate of metal complexes in cells to garner key mechanistic insights.
Collapse
Affiliation(s)
- Danielle van der Westhuizen
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg 2050, South Africa.
| | - Daniela I Bezuidenhout
- Laboratory of Inorganic Chemistry, Environmental and Chemical Engineering, University of Oulu, P. O. Box 3000, 90014 Oulu, Finland.
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg 2050, South Africa.
| |
Collapse
|
39
|
Hyun Kim J, Ofori S, Mertens RT, Parkin S, Awuah SG. Water-Soluble Gold(III)-Metformin Complex Alters Mitochondrial Bioenergetics in Breast Cancer Cells. ChemMedChem 2021; 16:3222-3230. [PMID: 34159760 PMCID: PMC8526394 DOI: 10.1002/cmdc.202100233] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/16/2021] [Indexed: 12/30/2022]
Abstract
Chemical control of mitochondrial dynamics and bioenergetics can unravel fundamental biological mechanisms and therapeutics for several diseases including, diabetes and cancer. We synthesized stable, water-soluble gold(III) complexes (Auraformin) supported by biguanide metformin or phenylmetformin for efficacious inhibition of mitochondrial respiration. The new compounds were characterized following the reaction of [C N]-cyclometalated gold(III) compounds with respective biguanides. Auraformin is solution stable in a physiologically relevant environment. We show that auraformin decreases mitochondrial respiration efficiently in comparison to the clinically used metformin by 100-fold. The compound displays significant mitochondrial uptake and induces antiproliferative activity in the micromolar range. Our results shed light on the development of new scaffolds as improved inhibitors of mitochondrial respiration.
Collapse
Affiliation(s)
- Jong Hyun Kim
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
| | - R Tyler Mertens
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
| | - Sean Parkin
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, 505 Rose Street, Lexington, KY 40506, USA
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone Street, Lexington, KY 40506, USA
| |
Collapse
|
40
|
Hu X, Li H, Ip TKY, Cheung YF, Koohi-Moghadam M, Wang H, Yang X, Tritton DN, Wang Y, Wang Y, Wang R, Ng KM, Naranmandura H, Tse EWC, Sun H. Arsenic trioxide targets Hsp60, triggering degradation of p53 and survivin. Chem Sci 2021; 12:10893-10900. [PMID: 34476069 PMCID: PMC8372542 DOI: 10.1039/d1sc03119h] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
The mechanisms of action of arsenic trioxide (ATO), a clinically used drug for the treatment of acute promyelocytic leukemia (APL), have been actively studied mainly through characterization of individual putative protein targets. There appear to be no studies at a system level. Herein, we integrate metalloproteomics through a newly developed organoarsenic probe, As-AC (C20H17AsN4O3S2) with quantitative proteomics, allowing 37 arsenic binding and 250 arsenic regulated proteins to be identified in NB4, a human APL cell line. Bioinformatics analysis reveals that ATO disrupts multiple physiological processes, in particular, chaperone-related protein folding and cellular response to stress. Furthermore, we discover heat shock protein 60 (Hsp60) as a vital target of ATO. Through biophysical and cell-based assays, we demonstrate that ATO binds to Hsp60, leading to abolishment of Hsp60 refolding capability. Significantly, the binding of ATO to Hsp60 disrupts the formation of Hsp60-p53 and Hsp60-survivin complexes, resulting in degradation of p53 and survivin. This study provides significant insights into the mechanism of action of ATO at a systemic perspective, and serves as guidance for the rational design of metal-based anticancer drugs. A highly selective organoarsenic fluorescent probe As-AC and quantitative proteomics were employed to track arsenic-binding and regulating proteins in live leukemia cells. Hsp60 was validated as a new target of ATO.![]()
Collapse
Affiliation(s)
- Xuqiao Hu
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Hongyan Li
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Tiffany Ka-Yan Ip
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Yam Fung Cheung
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Mohamad Koohi-Moghadam
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China .,Division of Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, University of Hong Kong Hong Kong SAR P. R. China
| | - Haibo Wang
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Xinming Yang
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Daniel N Tritton
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Yuchuan Wang
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Yi Wang
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Runming Wang
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Kwan-Ming Ng
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China .,Department of Chemistry, Shantou University Shantou Guangdong 515063 P. R. China
| | - Hua Naranmandura
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University Hangzhou P.R. China
| | - Eric Wai-Choi Tse
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital Hong Kong P. R. China
| | - Hongzhe Sun
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| |
Collapse
|
41
|
Pettenuzzo A, Vezzù K, Di Paolo ML, Fotopoulou E, Marchiò L, Via LD, Ronconi L. Design, physico-chemical characterization and in vitro biological activity of organogold(III) glycoconjugates. Dalton Trans 2021; 50:8963-8979. [PMID: 34110336 DOI: 10.1039/d1dt01100f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To develop new metal-based glycoconjugates as potential anticancer agents, four organometallic gold(iii)-dithiocarbamato glycoconjugates of the type [AuIII(2-Bnpy)(SSC-Inp-GlcN)](PF6) (2-Bnpy: 2-benzylpyridine; Inp: isonipecotic moiety; GlcN: amino-glucose scaffold; Au3-Au6) and the corresponding model non-glycosylated counterparts [AuIII(2-Bnpy)(SSC-Inp-R)](PF6) (R: OEt (Au1), NH2 (Au2)) have been generated and characterized by means of several analytical techniques (elemental analysis, FT-IR, 1H-/13C-NMR, ESI-MS, UV-Vis, X-ray crystallography). Their stability under physiologically-relevant conditions (PBS solution) and n-octanol/PBS distribution coefficient (D7.4) have also been evaluated. Gold(iii) glycoconjugates showed an antiproliferative effect against ovarian carcinoma A2780 cells, with GI50 values in the low micromolar range. Remarkably, their cell growth inhibitory effect increases upon the addition of a glucose transporter 1 (GLUT1) inhibitor, thus ruling out the involvement of GLUT1 in their transport inside the cell. Additional mechanistic studies have been carried out in A2780 cells, supporting the hypothesis of a facilitated diffusion mechanism (possibly mediated by glucose transporters other than GLUT1), and revealing their capability to act as topoisomerase I and II inhibitors and to disrupt mitochondrial membrane integrity, leading to the generation of ROS, thus resulting in the promotion of oxidative stress and, eventually, cell death.
Collapse
Affiliation(s)
- Andrea Pettenuzzo
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| | - Keti Vezzù
- University of Padova, Department of Industrial Engineering, Via F. Marzolo 8, 35131 Padova, Italy
| | - Maria Luisa Di Paolo
- University of Padova, Department of Molecular Medicine, Via G. Colombo 3, 35131 Padova, Italy
| | - Eirini Fotopoulou
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| | - Luciano Marchiò
- University of Parma, Department of Chemistry, Life Sciences and Environmental Sustainability, Parco Area delle Scienze 11/a, 43124 Parma, Italy
| | - Lisa Dalla Via
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, 35131 Padova, Italy.
| | - Luca Ronconi
- National University of Ireland Galway, School of Chemistry, University Road, H91 TK33 Galway, Co. Galway, Ireland.
| |
Collapse
|
42
|
N-Heterocyclic Carbene-Gold(I) Complexes Targeting Actin Polymerization. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11125626] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Transition metal complexes are attracting attention because of their various chemical and biological properties. In particular, the NHC-gold complexes represent a productive field of research in medicinal chemistry, mostly as anticancer tools, displaying a broad range of targets. In addition to the already known biological targets, recently, an important activity in the organization of the cell cytoskeleton was discovered. In this paper, we demonstrated that two NHC-gold complexes (namely AuL4 and AuL7) possessing good anticancer activity and multi-target properties, as stated in our previous studies, play a major role in regulating the actin polymerization, by the means of in silico and in vitro assays. Using immunofluorescence and direct enzymatic assays, we proved that both the complexes inhibited the actin polymerization reaction without promoting the depolymerization of actin filaments. Our outcomes may contribute toward deepening the knowledge of NHC-gold complexes, with the objective of producing more effective and safer drugs for treating cancer diseases.
Collapse
|
43
|
Churusova SG, Aleksanyan DV, Rybalkina EY, Susova OY, Peregudov AS, Brunova VV, Gutsul EI, Klemenkova ZS, Nelyubina YV, Glushko VN, Kozlov VA. Palladium(II) Pincer Complexes of Functionalized Amides with S-Modified Cysteine and Homocysteine Residues: Cytotoxic Activity and Different Aspects of Their Biological Effect on Living Cells. Inorg Chem 2021; 60:9880-9898. [PMID: 34130457 DOI: 10.1021/acs.inorgchem.1c01138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the search for potential new metal-based antitumor agents, two series of nonclassical palladium(II) pincer complexes based on functionalized amides with S-modified cysteine and homocysteine residues have been prepared and fully characterized by 1D and 2D NMR (1H, 13C, COSY, HMQC or HSQC, 1H-13C, and 1H-15N HMBC) and IR spectroscopy and, in some cases, X-ray diffraction. Most of the resulting complexes exhibit a high level of cytotoxic activity against several human cancer cell lines, including colon (HCT116), breast (MCF7), and prostate (PC3) cancers. Some of the compounds under consideration are also efficient in both native and doxorubicin-resistant transformed breast cells HBL100, suggesting the prospects for the creation of therapeutic agents based on the related compounds that would be able to overcome drug resistance. An analysis of different aspects of their biological effects on living cells has revealed a remarkable ability of the S-modified derivatives to induce cell apoptosis and efficient cellular uptake of their fluorescein-conjugated counterpart, confirming the high anticancer potential of Pd(II) pincer complexes derived from functionalized amides with S-donor amino acid pendant arms.
Collapse
Affiliation(s)
- Svetlana G Churusova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, ul. Vavilova 28, Moscow 119991, Russia
| | - Diana V Aleksanyan
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, ul. Vavilova 28, Moscow 119991, Russia
| | - Ekaterina Yu Rybalkina
- Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Kashirskoe sh. 23, Moscow 115478, Russia
| | - Olga Yu Susova
- Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Kashirskoe sh. 23, Moscow 115478, Russia
| | - Alexander S Peregudov
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, ul. Vavilova 28, Moscow 119991, Russia
| | - Valentina V Brunova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, ul. Vavilova 28, Moscow 119991, Russia
| | - Evgenii I Gutsul
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, ul. Vavilova 28, Moscow 119991, Russia
| | - Zinaida S Klemenkova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, ul. Vavilova 28, Moscow 119991, Russia
| | - Yulia V Nelyubina
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, ul. Vavilova 28, Moscow 119991, Russia
| | - Valentina N Glushko
- Institute of Chemical Reagents and High Purity Chemical Substances of the National Research Centre "Kurchatov Institute", Bogorodskii val 3, Moscow 107076, Russia
| | - Vladimir A Kozlov
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, ul. Vavilova 28, Moscow 119991, Russia
| |
Collapse
|
44
|
Wu S, Wu Z, Ge Q, Zheng X, Yang Z. Antitumor activity of tridentate pincer and related metal complexes. Org Biomol Chem 2021; 19:5254-5273. [PMID: 34059868 DOI: 10.1039/d1ob00577d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Pincer complexes featuring tunable tridentate ligand frameworks are one of the most actively studied classes of metal-based complexes. Currently, growing attention is devoted to the cytotoxicity of pincer and related metal complexes. The antiproliferative activity of numerous pincer complexes has been reported. Pincer tridentate ligand scaffolds show different coordination modes and offer multiple options for directed structural modifications. This review summarizes the significant progress in the research studies of the antitumor activity of pincer and related platinum(ii), gold(iii), palladium(ii), copper(ii), iron(iii), ruthenium(ii), nickel(ii) and some other metal complexes, in order to provide a reference for designing novel metal coordination drug candidates with promising antitumor activity.
Collapse
Affiliation(s)
- Shulei Wu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| | - Zaoduan Wu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| | - Qianyi Ge
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| | - Xing Zheng
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| | - Zehua Yang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| |
Collapse
|
45
|
Kim JH, Ofori S, Parkin S, Vekaria H, Sullivan PG, Awuah SG. Anticancer gold(iii)-bisphosphine complex alters the mitochondrial electron transport chain to induce in vivo tumor inhibition. Chem Sci 2021; 12:7467-7479. [PMID: 34163837 PMCID: PMC8171344 DOI: 10.1039/d1sc01418h] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/16/2021] [Indexed: 01/10/2023] Open
Abstract
Expanding the chemical diversity of metal complexes provides a robust platform to generate functional bioactive reagents. To access an excellent repository of metal-based compounds for probe/drug discovery, we capitalized on the rich chemistry of gold to create organometallic gold(iii) compounds by ligand tuning. We obtained novel organogold(iii) compounds bearing a 1,2-bis(diphenylphosphino)benzene ligand, providing structural diversity with optimal physiological stability. Biological evaluation of the lead compound AuPhos-89 demonstrates mitochondrial complex I-mediated alteration of the mitochondrial electron transport chain (ETC) to drive respiration and diminish cellular energy in the form of adenosine triphosphate (ATP). Mechanism-of-action efforts, RNA-Seq, quantitative proteomics, and NCI-60 screening reveal a highly potent anticancer agent that modulates mitochondrial ETC. AuPhos-89 inhibits the tumor growth of metastatic triple negative breast cancer and represents a new strategy to study the modulation of mitochondrial respiration for the treatment of aggressive cancer and other disease states where mitochondria play a pivotal role in the pathobiology.
Collapse
Affiliation(s)
- Jong Hyun Kim
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Sean Parkin
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Hemendra Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky USA
- Department of Neuroscience, University of Kentucky USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky USA
- Department of Neuroscience, University of Kentucky USA
- Lexington Veterans' Affairs Healthcare System USA
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky Lexington Kentucky 40536 USA
| |
Collapse
|
46
|
Platinum(II) N-heterocyclic carbene complexes arrest metastatic tumor growth. Proc Natl Acad Sci U S A 2021; 118:2025806118. [PMID: 33883283 DOI: 10.1073/pnas.2025806118] [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] [Indexed: 11/18/2022] Open
Abstract
Vimentin is a cytoskeletal intermediate filament protein that plays pivotal roles in tumor initiation, progression, and metastasis, and its overexpression in aggressive cancers predicted poor prognosis. Herein described is a highly effective antitumor and antimetastatic metal complex [PtII(C^N^N)(NHC2Bu)]PF6 (Pt1a; HC^N^N = 6-phenyl-2,2'-bipyridine; NHC= N-heterocyclic carbene) that engages vimentin via noncovalent binding interactions with a distinct orthogonal structural scaffold. Pt1a displays vimentin-binding affinity with a dissociation constant of 1.06 µM from surface plasmon resonance measurements and fits into a pocket between the coiled coils of the rod domain of vimentin with multiple hydrophobic interactions. It engages vimentin in cellulo, disrupts vimentin cytoskeleton, reduces vimentin expression in tumors, suppresses xenograft growth and metastasis in different mouse models, and is well tolerated, attributable to biotransformation to less toxic and renal-clearable platinum(II) species. Our studies uncovered the practical therapeutic potential of platinum(II)‒NHC complexes as effective targeted chemotherapy for combating metastatic and cisplatin-resistant cancers.
Collapse
|
47
|
Guarra F, Pratesi A, Gabbiani C, Biver T. A focus on the biological targets for coinage metal-NHCs as potential anticancer complexes. J Inorg Biochem 2021; 217:111355. [PMID: 33596529 DOI: 10.1016/j.jinorgbio.2021.111355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/27/2020] [Accepted: 01/08/2021] [Indexed: 12/11/2022]
Abstract
Metal complexes of N-heterocyclic carbene (NHC) ligands are the object of increasing attention for therapeutic purposes. Among the different metal centres, interest on Au-based compounds started with the application as anti-arthritis drugs. On the other hand, Ag(I) antimicrobial properties have been known for a long time. For Au(I)/Au(III)-NHC and Ag(I)-NHC anti-tumour and anti-proliferative properties have been quite recently demonstrated. In addition to these and as for Group 11, copper is a much less investigated metal centre, but a few papers underline its pharmacological potential. This review wants to focus on the different biological targets for these metal-based compounds. It is divided into chapters which are respectively devoted on: i) mitochondria and thiol oxidoreductase systems; ii) other relevant enzymes; iii) nucleic acids. Examples of representative coinage NHCs for each of the targets are provided together with significant references on recent advances on the topic. Moreover, a final comment summarises the aspects enlightened by each chapter and provides some hints to better understand the metal-NHCs mechanistic behaviour based on structure-activity relationships.
Collapse
Affiliation(s)
- Federica Guarra
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy.
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Tarita Biver
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy; Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy.
| |
Collapse
|
48
|
Neuditschko B, Legin AA, Baier D, Schintlmeister A, Reipert S, Wagner M, Keppler BK, Berger W, Meier‐Menches SM, Gerner C. Die Wechselwirkung mit ribosomalen Proteinen begleitet die Stressinduktion des Wirkstoffkandidaten BOLD-100/KP1339 im endoplasmatischen Retikulum. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 133:5121-5126. [PMID: 38505777 PMCID: PMC10947255 DOI: 10.1002/ange.202015962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 11/09/2022]
Abstract
AbstractDer metallhaltige Wirkstoff BOLD‐100/KP1339 zeigte bereits vielversprechende Resultate in verschiedenen In vitro‐ und In vivo‐Tumormodellen sowie in klinischen Studien. Der detaillierte Wirkmechanismus wurde jedoch noch nicht komplett aufgeklärt. Als entscheidende Wirkstoffeffekte kristallisierten sich kürzlich die Stressinduktion im endoplasmatischen Retikulum (ER) und die damit einhergehende Modulierung von HSPA5 (GRP78) heraus. Das spontane und stabile Addukt zwischen BOLD‐100 und menschlichem Serumalbumin wurde als Immobilisierungsstrategie ausgewählt, um einen chemoproteomischen Ansatz auszuführen, der die ribosomalen Proteine RPL10, RPL24 und den Transkriptionsfaktor GTF2I als potentielle Interaktoren dieser Ru(III)‐Verbindung identifizierten. Dieses Ergebnis wurde mit proteomischen und transkriptomischen Profiling‐Experimenten kombiniert, was die Interpretation einer ribosomalen Beeinträchtigung sowie der Induktion von ER‐Stress unterstützte. Die Bildung von Polyribosomen und begleitende ER‐Schwellungen in behandelten Krebszellen wurden zudem durch TEM‐Messungen bestätigt. Somit scheint eine direkte Wechselwirkung von BOLD‐100 mit ribosomalen Proteinen die ER‐Stressinduktion und die Modulierung von GRP78 in Krebszellen zu begleiten.
Collapse
Affiliation(s)
- Benjamin Neuditschko
- Institut für Anorganische ChemieFakultät für ChemieUniversität WienWähringer Str. 421090WienÖsterreich
- Institut für Analytische ChemieFakultät für ChemieUniversität WienWähringer Str. 381090WienÖsterreich
| | - Anton A. Legin
- Institut für Anorganische ChemieFakultät für ChemieUniversität WienWähringer Str. 421090WienÖsterreich
- Forschungsnetzwerk “Chemistry, Microbiology and Environmental Systems Science”Universität WienWähringer Str. 421090WienÖsterreich
| | - Dina Baier
- Institut für Anorganische ChemieFakultät für ChemieUniversität WienWähringer Str. 421090WienÖsterreich
- Institut für Krebsforschung und Comprehensive Cancer CenterUniversitätsklinik für Innere Medizin IMedizinische Universität WienBorschkegasse 8a1090WienÖsterreich
- Forschungscluster “Translational Cancer Therapy Research”Universität WienWähringer Str. 421090WienÖsterreich
| | - Arno Schintlmeister
- Forschungsnetzwerk “Chemistry, Microbiology and Environmental Systems Science”Universität WienWähringer Str. 421090WienÖsterreich
- Großgeräteeinrichtung für Umwelt- und Isotopen-MassenspektrometrieZentrum für Mikrobiologie und UmweltsystemwissenschaftUniversität WienAlthanstr. 141090WienÖsterreich
| | - Siegfried Reipert
- Core Facility für Cell Imaging und UltrastrukturforschungAlthanstr. 141090WienÖsterreich
| | - Michael Wagner
- Forschungsnetzwerk “Chemistry, Microbiology and Environmental Systems Science”Universität WienWähringer Str. 421090WienÖsterreich
- Großgeräteeinrichtung für Umwelt- und Isotopen-MassenspektrometrieZentrum für Mikrobiologie und UmweltsystemwissenschaftUniversität WienAlthanstr. 141090WienÖsterreich
| | - Bernhard K. Keppler
- Institut für Anorganische ChemieFakultät für ChemieUniversität WienWähringer Str. 421090WienÖsterreich
- Forschungsnetzwerk “Chemistry, Microbiology and Environmental Systems Science”Universität WienWähringer Str. 421090WienÖsterreich
- Forschungscluster “Translational Cancer Therapy Research”Universität WienWähringer Str. 421090WienÖsterreich
| | - Walter Berger
- Institut für Krebsforschung und Comprehensive Cancer CenterUniversitätsklinik für Innere Medizin IMedizinische Universität WienBorschkegasse 8a1090WienÖsterreich
- Forschungscluster “Translational Cancer Therapy Research”Universität WienWähringer Str. 421090WienÖsterreich
| | - Samuel M. Meier‐Menches
- Institut für Analytische ChemieFakultät für ChemieUniversität WienWähringer Str. 381090WienÖsterreich
- Forschungscluster “Translational Cancer Therapy Research”Universität WienWähringer Str. 421090WienÖsterreich
| | - Christopher Gerner
- Institut für Analytische ChemieFakultät für ChemieUniversität WienWähringer Str. 381090WienÖsterreich
- Joint Metabolome FacilityUniversität Wien und Medizinische Universität WienWähringer Str. 381090WienÖsterreich
| |
Collapse
|
49
|
Neuditschko B, Legin AA, Baier D, Schintlmeister A, Reipert S, Wagner M, Keppler BK, Berger W, Meier‐Menches SM, Gerner C. Interaction with Ribosomal Proteins Accompanies Stress Induction of the Anticancer Metallodrug BOLD-100/KP1339 in the Endoplasmic Reticulum. Angew Chem Int Ed Engl 2021; 60:5063-5068. [PMID: 33369073 PMCID: PMC7986094 DOI: 10.1002/anie.202015962] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 02/06/2023]
Abstract
The ruthenium-based anticancer agent BOLD-100/KP1339 has shown promising results in several in vitro and in vivo tumour models as well as in early clinical trials. However, its mode of action remains to be fully elucidated. Recent evidence identified stress induction in the endoplasmic reticulum (ER) and concomitant down-modulation of HSPA5 (GRP78) as key drug effects. By exploiting the naturally formed adduct between BOLD-100 and human serum albumin as an immobilization strategy, we were able to perform target-profiling experiments that revealed the ribosomal proteins RPL10, RPL24, and the transcription factor GTF2I as potential interactors of this ruthenium(III) anticancer agent. Integrating these findings with proteomic profiling and transcriptomic experiments supported ribosomal disturbance and concomitant induction of ER stress. The formation of polyribosomes and ER swelling of treated cancer cells revealed by TEM validated this finding. Thus, the direct interaction of BOLD-100 with ribosomal proteins seems to accompany ER stress-induction and modulation of GRP78 in cancer cells.
Collapse
Affiliation(s)
- Benjamin Neuditschko
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 421090ViennaAustria
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 381090ViennaAustria
| | - Anton A. Legin
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 421090ViennaAustria
- Research Network “Chemistry, Microbiology and Environmental Systems Science”University of ViennaWähringer Str. 421090ViennaAustria
| | - Dina Baier
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 421090ViennaAustria
- Institute of Cancer Research and Comprehensive Cancer CenterDepartment of Medicine IMedical University of ViennaBorschkegasse 8a1090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”University of ViennaWaehringer Str. 421090ViennaAustria
| | - Arno Schintlmeister
- Research Network “Chemistry, Microbiology and Environmental Systems Science”University of ViennaWähringer Str. 421090ViennaAustria
- Large-Instrument Facility for Environmental and Isotope Mass SpectrometryCentre for Microbiology and Environmental Systems ScienceUniversity of ViennaAlthanstr. 141090ViennaAustria
| | - Siegfried Reipert
- Core Facility Cell Imaging and Ultrastructure ResearchAlthanstr. 141090ViennaAustria
| | - Michael Wagner
- Research Network “Chemistry, Microbiology and Environmental Systems Science”University of ViennaWähringer Str. 421090ViennaAustria
- Large-Instrument Facility for Environmental and Isotope Mass SpectrometryCentre for Microbiology and Environmental Systems ScienceUniversity of ViennaAlthanstr. 141090ViennaAustria
| | - Bernhard K. Keppler
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 421090ViennaAustria
- Research Network “Chemistry, Microbiology and Environmental Systems Science”University of ViennaWähringer Str. 421090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”University of ViennaWaehringer Str. 421090ViennaAustria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer CenterDepartment of Medicine IMedical University of ViennaBorschkegasse 8a1090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”University of ViennaWaehringer Str. 421090ViennaAustria
| | - Samuel M. Meier‐Menches
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 381090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”University of ViennaWaehringer Str. 421090ViennaAustria
| | - Christopher Gerner
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 381090ViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University of ViennaWaehringer Str. 381090ViennaAustria
| |
Collapse
|
50
|
Affiliation(s)
- Sina Witzel
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
| | - A. Stephen K. Hashmi
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120 Heidelberg, Germany
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Jin Xie
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|