1
|
Xu S, Wu X, Zhu J, Wu Q, Gao L, Yang F, Zhang Z. Research Progress of Endoplasmic Reticulum Targeting Metal Complexes in Cancer Therapy. Drug Dev Res 2024; 85:e70027. [PMID: 39676587 DOI: 10.1002/ddr.70027] [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: 08/03/2024] [Revised: 10/25/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
The development of anticancer drugs that target different organelles has received extensive attention due to the characteristics of cancer recurrence, metastasis, and drug resistance. The endoplasmic reticulum (ER) is an important structure within the cell that is primarily responsible for protein synthesis, folding, modification, and transport and plays a crucial role in cell function and health. ER stress activation induces cancer cell apoptosis. New anticancer drugs with different anticancer mechanisms and selectivity can be designed because of redox activity, composition diversity, and metal complexes structure regulation. Over the past few decades, dozens of metal complexes have killed cancer cells through ER stress, showing powerful tumor-suppressive effects. This review summarizes the progress of research on anticancer metallic drugs that induce ER stress over the past few years, which is expected to bring more breakthroughs in the field of medicine and life science.
Collapse
Affiliation(s)
- Shihang Xu
- School Hospital, Guangxi Normal University, Guilin, Guangxi, P.R. China
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Xiaoling Wu
- School Hospital, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Jia Zhu
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Qiuming Wu
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Lijuan Gao
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| | - Zhenlei Zhang
- School Hospital, Guangxi Normal University, Guilin, Guangxi, P.R. China
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, P.R. China
| |
Collapse
|
2
|
Tu Y, Gong J, Mou J, Jiang H, Zhao H, Gao J. Strategies for the development of stimuli-responsive small molecule prodrugs for cancer treatment. Front Pharmacol 2024; 15:1434137. [PMID: 39144632 PMCID: PMC11322083 DOI: 10.3389/fphar.2024.1434137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
Approved anticancer drugs typically face challenges due to their narrow therapeutic window, primarily because of high systemic toxicity and limited selectivity for tumors. Prodrugs are initially inactive drug molecules designed to undergo specific chemical modifications. These modifications render the drugs inactive until they encounter specific conditions or biomarkers in vivo, at which point they are converted into active drug molecules. This thoughtful design significantly improves the efficacy of anticancer drug delivery by enhancing tumor specificity and minimizing off-target effects. Recent advancements in prodrug design have focused on integrating these strategies with delivery systems like liposomes, micelles, and polymerosomes to further improve targeting and reduce side effects. This review outlines strategies for designing stimuli-responsive small molecule prodrugs focused on cancer treatment, emphasizing their chemical structures and the mechanisms controlling drug release. By providing a comprehensive overview, we aim to highlight the potential of these innovative approaches to revolutionize cancer therapy.
Collapse
Affiliation(s)
- Yuxuan Tu
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jianbao Gong
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao Municipal Hospital, Qingdao, China
| | - Jing Mou
- Department of Neonatology, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Hongfei Jiang
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Haibo Zhao
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jiake Gao
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Gomez-Lopez S, Serrano R, Cohen B, Martinez-Argudo I, Lopez-Sanz L, Guadamillas MC, Calero R, Ruiz MJ. Novel Titanocene Y derivative with albumin affinity exhibits improved anticancer activity against platinum resistant cells. J Inorg Biochem 2024; 254:112520. [PMID: 38460481 DOI: 10.1016/j.jinorgbio.2024.112520] [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: 12/21/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
The antitumor activity of Ti(IV)-based compounds put them in the spotlight for cancer treatment in the past, but their lack of stability in vivo due to a high rate of hydrolysis has hindered their development as antitumor drugs. As a possible solution for this problem, we have reported a synthesis strategy through which we combined a titanocene fragment, a tridentate ligand, and a long aliphatic chain. This strategy allowed us to generate a titanium compound (Myr-Ti) capable of interacting with albumin, highly stable in water and with cytotoxic activity in tumor cells[1]. Following a similar strategy, now we report the synthesis of a new compound (Myr-TiY) derived from titanocene Y that shows antitumoral activity in a cisplatin resistant model with a 50% inhibitory concentration (IC50) of 41-76 μM. This new compound shows high stability and a strong interaction with human serum albumin. Myr-TiY has a significant antiproliferative and proapoptotic effect on the tested cancer cells and shows potential tumor selectivity when assayed in non-tumor human epithelial cells being more selective (1.3-3.8 times) for tumor cells than cisplatin. These results lead us to think that the described synthesis strategy could be useful to generate compounds for the treatment of both cisplatin-sensitive and cisplatin-resistant cancers.
Collapse
Affiliation(s)
- Sergio Gomez-Lopez
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain
| | - Rosario Serrano
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain; Departamento de Química Orgánica, Inorgánica y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain
| | - Boiko Cohen
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain; Departamento de Química Física, Universidad de Castilla-La Mancha, 45071 Toledo, Spain; INAMOL, Universidad de Castilla-La Mancha, 45071 Toledo, Spain
| | - Isabel Martinez-Argudo
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain; Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, 45071 Toledo, Spain
| | - Laura Lopez-Sanz
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain
| | - Marta Carmen Guadamillas
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain; Departamento de Ciencia y Tecnología Agroforestal y Genética, Universidad de Castilla-La Mancha, 45071 Toledo, Spain
| | - Raul Calero
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain; Departamento de Química Orgánica, Inorgánica y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain
| | - Maria Jose Ruiz
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain; Departamento de Química Orgánica, Inorgánica y Bioquímica, Universidad de Castilla-La Mancha, 45071 Toledo, Spain; INAMOL, Universidad de Castilla-La Mancha, 45071 Toledo, Spain
| |
Collapse
|
4
|
Thanigachalam S, Pathak M. Bioactive O^N^O^ Schiff base appended homoleptic titanium(iv) complexes: DFT, BSA/CT-DNA interactions, molecular docking and antitumor activity against HeLa and A549 cell lines. RSC Adv 2024; 14:13062-13082. [PMID: 38655487 PMCID: PMC11034360 DOI: 10.1039/d3ra08574k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/06/2024] [Indexed: 04/26/2024] Open
Abstract
Five new homoleptic derivatives of titanium(iv) have been developed and characterized by physicochemical techniques. Metal complexes, TiH2L1 [(C38H26N6O4)Ti], TiH2L2 [(C38H24F2N6O4)Ti], TiH2L3 [(C38H24Cl2N6O4)Ti], TiH2L4 [(C38H24Br2N6O4)Ti] and TiH2L5 [(C38H24N8O8)Ti], were obtained by treating Ti(OPri)4 with appropriate ONO ligands (H2L1-H2L5) in anhydrous THF as solvent. The electronic structures and properties of titanium(iv) complexes (TiH2L1-TiH2L5) and ligands (H2L1-H2L5) were examined by DFT studies. The stability of all synthesized derivatives was assessed by a UV-visible technique using 10% DMSO, GSH medium and n-octanol/water systems. The binding interactions of BSA and CT-DNA with respective titanium(iv) complexes were successfully evaluated by employing UV-visible absorption, fluorescence, circular dichroism (CD) techniques and docking studies. The in vitro cytotoxicity of TiH2L2, TiH2L3 and TiH2L4 complexes was assessed against HeLa (human epithelioid cervical cancer cells) and A549 (lung carcinoma) cell lines. The IC50 values of TiH2L2, TiH2L3 and TiH2L4 were observed to be 28.8, 14.7 and 31.2 μg mL-1 for the HeLa cell line and 38.2, 32.9 and 67.78 μg mL-1 for A549 cells, respectively. Complex TiH2L3 exhibited remarkably induced cell cycle arrest in the G1 phase and 77.99% ROS production selectivity in the HeLa cell line.
Collapse
Affiliation(s)
- Sathish Thanigachalam
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology Vellore 632014 Tamilnadu India
| | - Madhvesh Pathak
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology Vellore 632014 Tamilnadu India
| |
Collapse
|
5
|
Musa M, Abid M, Bradshaw TD, Boocock DJ, Coveney C, Argent SP, Woodward S. Probing the Mechanism of Action of Bis(phenolato) Amine (ONO Donor Set) Titanium(IV) Anticancer Agents. J Med Chem 2024; 67:2732-2744. [PMID: 38331433 PMCID: PMC10895680 DOI: 10.1021/acs.jmedchem.3c01874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024]
Abstract
The need for anticancer therapies that overcome metallodrug resistance while minimizing adverse toxicities is targeted, herein, using titanium coordination complexes. Octahedral titanium(IV) trans,mer-[Ti{R1N(CH2-2-MeO-4-R1-C6H2)2}2] [R1 = Et, allyl, n-Pr, CHO, F, CH2(morpholino), the latter from the formyl derivative; R2 = Me, Et; not all combinations] are attained from Mannich reactions of commercial 2-methoxyphenols (27-74% overall yield, 2 steps). These crystalline (four X-ray structures) Ti(IV)-complexes are active against MCF-7, HCT-116, HT-29, PANC-1, and MDA-MB-468 cancer cell lines (GI50 = 0.5-38 μM). Their activity and cancer selectivity (vs nontumor MRC-5 cells) typically exceeds that of cisplatin (up to 16-fold). Proteomic analysis (in MCF-7) supported by other studies (G2/M cell cycle arrest, ROS generation, γH2AX production, caspase activation, annexin positivity, western blot, and kinase screens in MCF-7 and HCT-116) suggest apoptosis elicited by more than one mechanism of action. Comparison of these data to the modes of action proposed for salan Ti(IV) complexes is made.
Collapse
Affiliation(s)
- Mustapha Musa
- GSK
Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Triumph Road, Nottingham NG7 2TU, U.K.
| | - Mohammed Abid
- Department
of Chemistry, College of Science, University
of Anbar, Anbarshire 31001, Iraq
| | - Tracey D. Bradshaw
- BDI,
School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - David J. Boocock
- School
of Science and Technology, Nottingham Trent
University, Clifton, Nottingham NG11 8NS, U.K.
| | - Clare Coveney
- School
of Science and Technology, Nottingham Trent
University, Clifton, Nottingham NG11 8NS, U.K.
| | - Stephen P. Argent
- School
of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Simon Woodward
- GSK
Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Triumph Road, Nottingham NG7 2TU, U.K.
| |
Collapse
|
6
|
Kostova I. Anticancer Metallocenes and Metal Complexes of Transition Elements from Groups 4 to 7. Molecules 2024; 29:824. [PMID: 38398576 PMCID: PMC10891901 DOI: 10.3390/molecules29040824] [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: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
With the progression in the field of bioinorganic chemistry, the role of transition metal complexes as the most widely used therapeutics is becoming a more and more attractive research area. The complexes of transition metals possess a great variety of attractive pharmacological properties, including anticancer, anti-inflammatory, antioxidant, anti-infective, etc., activities. Transition metal complexes have proven to be potential alternatives to biologically active organic compounds, especially as antitumor agents. The performance of metal coordination compounds in living systems is anticipated to differ generally from the action of non-metal-containing drugs and may offer unique diagnostic and/or therapeutic opportunities. In this review, the rapid development and application of metallocenes and metal complexes of elements from Groups 4 to 7 in cancer diagnostics and therapy have been summarized. Most of the heavy metals discussed in the current review are newly discovered metals. That is why the use of their metal-based compounds has attracted a lot of attention concerning their organometallic and coordination chemistry. All of this imposes more systematic studies on their biological activity, biocompatibility, and toxicity and presupposes further investigations.
Collapse
Affiliation(s)
- Irena Kostova
- Department of Chemistry, Faculty of Pharmacy, Medical University-Sofia, 1000 Sofia, Bulgaria
| |
Collapse
|
7
|
Kumar N, Kaushal R, Awasthi P. A Comprehensive Review on the Development of Titanium Complexes as Cytotoxic Agents. Curr Top Med Chem 2024; 24:2117-2128. [PMID: 39108106 DOI: 10.2174/0115680266317770240718080512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 10/22/2024]
Abstract
After the discovery of cis-platin, the first metal-based anticancer drugs, budotitane, and titanocene dichloride entered clinical trials. These two classes of complexes were effective against those cell lines that are resistant to cis-platin and other platinum-based drugs. However, the main limitation of these complexes is their low hydrolytic stability. After these two classes, a third generation titanium based complex, i.e. diaminebis(phenolato)bis(alkoxo) titanium(IV), was invented, which showed more hydrolytic stability and high cytotoxicity than budotitane and titanocene dichloride. The Hydrolytic stability of complexes plays an important role in cytotoxicity. Earlier research showed that hydrolytically less stable complexes decompose rapidly into non-bioavailable moiety and become inactive. The mechanism of Ti(IV) complexes of diaminebis(phenolato) bis(alkoxo) is under investigation and is presumed to involve Endoplasmic Reticulum (ER) stress, which leads to apoptosis. The proposed mechanism involves the removal of ligands from the titanium complex and the binding of the Ti center to transferrin protein and its release inside the cell. Also, the structure of the ligand plays a key role in the cytotoxicity of complexes; as the bulkiness of the ligand increased, the cytotoxic nature of complexes decreased.
Collapse
Affiliation(s)
- Nitesh Kumar
- Department of Chemistry, Government College Jhandutta, District Bilaspur, Himachal Pradesh, 174031, India
| | - Raj Kaushal
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh, 177005, India
| | - Pamita Awasthi
- Department of Chemistry, National Institute of Technology, Hamirpur, Himachal Pradesh, 177005, India
| |
Collapse
|
8
|
Hayet S, Ghrayeb M, Azulay DN, Shpilt Z, Tshuva EY, Chai L. Titanium complexes affect Bacillus subtilis biofilm formation. RSC Med Chem 2023; 14:983-991. [PMID: 37252093 PMCID: PMC10211322 DOI: 10.1039/d3md00075c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/01/2023] [Indexed: 05/31/2023] Open
Abstract
Biofilms are surface or interface-associated communities of bacterial cells, embedded in a self-secreted extracellular matrix (ECM). Cells in biofilms are 100-1000 times more resistant to antibiotic treatment relative to planktonic cells due to various reasons, including the ECM acting as a diffusion barrier to antibiotic molecules, the presence of persister cells that divide slowly and are less susceptible to cell-wall targeting drugs, and the activation of efflux pumps in response to antibiotic stress. In this study we tested the effect of two titanium(iv) complexes that have been previously reported as potent and non-toxic anticancer chemotherapeutic agents on Bacillus subtilis cells in culture and in biofilm forming conditions. The Ti(iv) complexes tested, a hexacoordinate diaminobis(phenolato)-bis(alkoxo) complex (phenolaTi) and a bis(isopropoxo) complex of a diaminobis(phenolato) "salan"-type ligand (salanTi), did not affect the growth rate of cells in shaken cultures, however they did affect biofilm formation. Surprisingly, while phenolaTi inhibited biofilm formation, the presence of salanTi induced the formation of more mechanically robust biofilms. Optical microscopy images of biofilm samples in the absence and presence of Ti(iv) complexes suggest that Ti(iv) complexes affect cell-cell and/or cell-matrix adhesion, and that these are interfered with phenolaTi and enhanced by salanTi. Our results highlight the possible effect of Ti(iv) complexes on bacterial biofilms, which is gaining interest in light of the emerging relations between bacteria and cancerous tumors.
Collapse
Affiliation(s)
- Shahar Hayet
- Institute of Chemistry, The Hebrew University of Jerusalem Edmond J. Safra Campus Jerusalem 91904 Israel +972 2 5660425 +972 2 6586084 +972 2 6585303
- The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem Jerusalem Israel
| | - Mnar Ghrayeb
- Institute of Chemistry, The Hebrew University of Jerusalem Edmond J. Safra Campus Jerusalem 91904 Israel +972 2 5660425 +972 2 6586084 +972 2 6585303
- The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem Jerusalem Israel
| | - David N Azulay
- Institute of Chemistry, The Hebrew University of Jerusalem Edmond J. Safra Campus Jerusalem 91904 Israel +972 2 5660425 +972 2 6586084 +972 2 6585303
- The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem Jerusalem Israel
| | - Zohar Shpilt
- Institute of Chemistry, The Hebrew University of Jerusalem Edmond J. Safra Campus Jerusalem 91904 Israel +972 2 5660425 +972 2 6586084 +972 2 6585303
| | - Edit Y Tshuva
- Institute of Chemistry, The Hebrew University of Jerusalem Edmond J. Safra Campus Jerusalem 91904 Israel +972 2 5660425 +972 2 6586084 +972 2 6585303
| | - Liraz Chai
- Institute of Chemistry, The Hebrew University of Jerusalem Edmond J. Safra Campus Jerusalem 91904 Israel +972 2 5660425 +972 2 6586084 +972 2 6585303
- The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem Jerusalem Israel
| |
Collapse
|
9
|
Shpilt Z, Melamed-Book N, Tshuva EY. An anticancer Ti(IV) complex increases mitochondrial reactive oxygen species levels in relation with hypoxia and endoplasmic-reticulum stress: A distinct non DNA-related mechanism. J Inorg Biochem 2023; 243:112197. [PMID: 36963201 DOI: 10.1016/j.jinorgbio.2023.112197] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 03/26/2023]
Abstract
PhenolaTi is a promising Ti(IV) anticancer complex, with high stability and cytotoxicity, without notable toxic side-effects. Its cellular mechanism was proposed to relate to ER stress. Herein, we investigated the downstream effects of this mode of action in two cancer cell lines: ovarian carcinoma A2780 and cervical adenocarcinoma HeLa. First, although Ti(IV) is a non-redox metal, the formation of mitochondrial reactive oxygen species (ROS) was detected with live-cell imaging. Then, we inspected the effect of the mitochondrial ROS on cytotoxicity, using two methods: (a) addition of compounds that either elevate or reduce the mitochondrial glutathione concentration, thus affecting the oxidative state of the cells; and (b) scavenging mitochondrial ROS. Unlike the results observed for cisplatin, neither method influenced the cytotoxicity of phenolaTi, implying that ROS formation was a mere side effect of its activity. Additionally, live cell imaging displayed the hypoxia induced by phenolaTi, which can be associated with ROS formation. Overall, the results support the notion that ER-stress is the main cellular mechanism of phenolaTi, leading to hypoxia and mitochondrial ROS. The distinct mechanism of phenolaTi, which is different from that of cisplatin, combined with its stability and favorable anticancer properties, altogether make it a strong chemotherapeutic drug candidate.
Collapse
Affiliation(s)
- Zohar Shpilt
- Institute of Chemistry, the Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Naomi Melamed-Book
- The Bio-Imaging Unit, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Edit Y Tshuva
- Institute of Chemistry, the Hebrew University of Jerusalem, Jerusalem 9190401, Israel..
| |
Collapse
|
10
|
Shpilt Z, Tshuva EY. Stable, Cytotoxic, and Fluorescent Ti(IV) Phenolato Complexes – Synthesis, Characterization, and Potential Use in Live Cell Imaging. INORG CHEM COMMUN 2023. [DOI: 10.1016/j.inoche.2023.110660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
11
|
Han HH, Wang HM, Jangili P, Li M, Wu L, Zang Y, Sedgwick AC, Li J, He XP, James TD, Kim JS. The design of small-molecule prodrugs and activatable phototherapeutics for cancer therapy. Chem Soc Rev 2023; 52:879-920. [PMID: 36637396 DOI: 10.1039/d2cs00673a] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cancer remains as one of the most significant health problems, with approximately 19 million people diagnosed worldwide each year. Chemotherapy is a routinely used method to treat cancer patients. However, current treatment options lack the appropriate selectivity for cancer cells, are prone to resistance mechanisms, and are plagued with dose-limiting toxicities. As such, researchers have devoted their attention to developing prodrug-based strategies that have the potential to overcome these limitations. This tutorial review highlights recently developed prodrug strategies for cancer therapy. Prodrug examples that provide an integrated diagnostic (fluorescent, photoacoustic, and magnetic resonance imaging) response, which are referred to as theranostics, are also discussed. Owing to the non-invasive nature of light (and X-rays), we have discussed external excitation prodrug strategies as well as examples of activatable photosensitizers that enhance the precision of photodynamic therapy/photothermal therapy. Activatable photosensitizers/photothermal agents can be seen as analogous to prodrugs, with their phototherapeutic properties at a specific wavelength activated in the presence of disease-related biomarkers. We discuss each design strategy and illustrate the importance of targeting biomarkers specific to the tumour microenvironment and biomarkers that are known to be overexpressed within cancer cells. Moreover, we discuss the advantages of each approach and highlight their inherent limitations. We hope in doing so, the reader will appreciate the current challenges and available opportunities in the field and inspire subsequent generations to pursue this crucial area of cancer research.
Collapse
Affiliation(s)
- Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, P. R. China. .,State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, P. R. China
| | - Han-Min Wang
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Paramesh Jangili
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Mingle Li
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Luling Wu
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK.
| | - Yi Zang
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,Lingang laboratory, Shanghai 201203, China
| | - Adam C Sedgwick
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, OX1 3TA, UK.
| | - Jia Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, P. R. China. .,The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China.,National Center for Liver Cancer, Shanghai 200438, China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK. .,School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
12
|
Scarpi-Luttenauer M, Galentino K, Orvain C, Cecchini M, Gaiddon C, Mobian P. TiO4N2 complexes formed with 1,10-phenanthroline ligands containing a donor-acceptor hydrogen bond site: synthesis, cytotoxicity and docking experiments. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.121036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
13
|
Binding of the anticancer Ti(IV) complex phenolaTi to serum proteins: Thermodynamic and kinetic aspects. J Inorg Biochem 2022; 232:111817. [DOI: 10.1016/j.jinorgbio.2022.111817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 11/20/2022]
|
14
|
Yim J, Park SB. Label-Free Target Identification Reveals the Anticancer Mechanism of a Rhenium Isonitrile Complex. Front Chem 2022; 10:850638. [PMID: 35372261 PMCID: PMC8964423 DOI: 10.3389/fchem.2022.850638] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/25/2022] [Indexed: 01/21/2023] Open
Abstract
Elucidation of the molecular mechanism of therapeutic agents and potential candidates is in high demand. Interestingly, rhenium-based complexes have shown a highly selective anticancer effect, only on cancer cells, unlike platinum-based drugs, such as cisplatin and carboplatin. These differences might be attributed to their different molecular targets. We confirmed that the target of tricarbonyl rhenium isonitrile polypyridyl (TRIP) complex is a protein, not DNA, using ICP-MS analysis and identified heat shock protein 60 (HSP60) as its target protein using a label-free target identification method. The subsequent biological evaluation revealed that TRIP directly inhibits the chaperone function of HSP60 and induces the accumulation of misfolded proteins in mitochondria, thereby leading to the activation of mitochondrial unfolded protein response (mtUPR)-mediated JNK2/AP-1/CHOP apoptotic pathway.
Collapse
Affiliation(s)
- Junhyeong Yim
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, South Korea
| | - Seung Bum Park
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, South Korea
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, South Korea
- *Correspondence: Seung Bum Park,
| |
Collapse
|
15
|
Rousselle B, Massot A, Privat M, Dondaine L, Trommenschlager A, Bouyer F, Bayardon J, Ghiringhelli F, Bettaieb A, Goze C, Paul C, Malacea-Kabbara R, Bodio E. Conception and evaluation of fluorescent phosphine-gold complexes: from synthesis to in vivo investigations. ChemMedChem 2022; 17:e202100773. [PMID: 35254001 DOI: 10.1002/cmdc.202100773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/02/2022] [Indexed: 11/11/2022]
Abstract
A phosphine gold(I) and phosphine-phosphonium gold(I) complexes bearing a fluorescent coumarin moiety were synthesized and characterized. Both complexes displayed interesting photophysical properties: good molar absorption coefficient, good quantum yield of fluorescence, and ability to be tracked in vitro thanks to two-photon imaging. Their in vitro and in vivo biological properties were evaluated onto cancer cell lines both human and murine and into CT26 tumor-bearing BALB/c mice. They displayed moderate to strong antiproliferative properties and the phosphine-phosphonium gold(I) complex induced significant in vivo anti-cancer effect.
Collapse
Affiliation(s)
- Benjamin Rousselle
- Université Bourgogne Franche-Comté: Universite Bourgogne Franche-Comte, ICMUB, FRANCE
| | - Aurélie Massot
- EPHE PSL: Ecole Pratique des Hautes Etudes, LIIC, FRANCE
| | - Malorie Privat
- Université Bourgogne Franche-Comté: Universite Bourgogne Franche-Comte, ICMUB and LIIC, FRANCE
| | - Lucile Dondaine
- Université Bourgogne Franche-Comté: Universite Bourgogne Franche-Comte, ICMUB and LIIC, FRANCE
| | | | - Florence Bouyer
- Université Bourgogne Franche-Comté: Universite Bourgogne Franche-Comte, INSERM 1231, FRANCE
| | - Jérôme Bayardon
- Université Bourgogne Franche-Comté: Universite Bourgogne Franche-Comte, ICMUB, FRANCE
| | - François Ghiringhelli
- Université Bourgogne Franche-Comté: Universite Bourgogne Franche-Comte, INSERM UMR 1231, FRANCE
| | - Ali Bettaieb
- EPHE PSL: Ecole Pratique des Hautes Etudes, LIIC, FRANCE
| | - Christine Goze
- Université Bourgogne Franche-Comté: Universite Bourgogne Franche-Comte, ICMUB, FRANCE
| | - Catherine Paul
- EPHE PSL: Ecole Pratique des Hautes Etudes, LIIC, FRANCE
| | | | - Ewen Bodio
- Burgundy University, Institut de Chimie Moleculaire de l'Universite de Bourgogne - UMR CNRS 6302, 9 avenue Alain Savary, BP 47870, 21078, Dijon, FRANCE
| |
Collapse
|
16
|
|
17
|
Yousuf I, Bashir M, Arjmand F, Tabassum S. Advancement of metal compounds as therapeutic and diagnostic metallodrugs: Current frontiers and future perspectives. Coord Chem Rev 2021; 445:214104. [DOI: 10.1016/j.ccr.2021.214104] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
18
|
Nahari G, Tshuva EY. Synthesis of asymmetrical diaminobis(alkoxo)-bisphenol compounds and their C 1-symmetrical mono-ligated titanium(iv) complexes as highly stable highly active antitumor compounds. Dalton Trans 2021; 50:6423-6426. [PMID: 33949509 PMCID: PMC8130176 DOI: 10.1039/d1dt00219h] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/18/2021] [Indexed: 11/26/2022]
Abstract
Asymmetrical 2,2'-((ethane-1,2-diylbis((2-hydroxyethyl)azanediyl))bis(methylene))diphenol substituted compounds and their C1-symmetrical diaminobis(phenolato)-bis(alkoxo) titanium(iv) complexes were synthesized, with one symmetrical analogue. X-ray crystallography corroborated tight ligand binding. Different substitutions on the two aromatic rings enabled fine-tuning of the complex properties, giving enhanced solubility, high anticancer activity (IC50 < 4 μM), and significant hydrolytic stability.
Collapse
Affiliation(s)
- Gilad Nahari
- The Institute of Chemistry, The Hebrew University of JerusalemJerusalem 9190401Israel
| | - Edit Y. Tshuva
- The Institute of Chemistry, The Hebrew University of JerusalemJerusalem 9190401Israel
| |
Collapse
|
19
|
Nahari G, Hoffman RE, Tshuva EY. From medium to endoplasmic reticulum: Tracing anticancer phenolato titanium(IV) complex by 19F NMR detection. J Inorg Biochem 2021; 221:111492. [PMID: 34051630 DOI: 10.1016/j.jinorgbio.2021.111492] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/22/2021] [Accepted: 05/12/2021] [Indexed: 01/12/2023]
Abstract
Titanium(IV) complexes of diaminobis(phenolato)-bis(alkoxo) ligands are promising anticancer drugs, showing marked in-vivo efficacy with no toxic side-effects in mice, hence, it is of interest to elucidate their mechanism of action. Herein, we employed a fluoro-substituted derivative, FenolaTi, for mechanistic analysis of the active species and its cellular target by quantitative 19F NMR detection to reveal its biodistribution and reactivity in extracellular and intracellular matrices. Upon administration to the serum-containing medium, FenolaTi interacted with bovine serum albumin. 20 h post administration, the cellular accumulation of FenolaTi derivatives was estimated as 37% of the administered compound, in a concentration three orders-of-magnitude higher than the administered dose, implying that active membrane transportation facilitates cellular penetration. An additional 19% of the administered dose that was detected in the extracellular environment had originated from post-apoptotic cells. In the cell, interaction with cellular proteins was detected. Although some intact Ti(IV) complex localized in the nucleus, no signals for isolated DNA fractions were detected and no reactivity with nuclear proteins was observed. Interestingly, higher accumulation of FenolaTi-derived compounds in the endoplasmic reticulum (ER) and interaction with proteins therein were detected, supporting the role of the ER as a possible target for cytotoxic bis(phenolato)-bis(alkoxo) Ti(IV) complexes.
Collapse
Affiliation(s)
- Gilad Nahari
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Roy E Hoffman
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Edit Y Tshuva
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| |
Collapse
|
20
|
Zhao T, Wang P, Ji M, Li S, Yang M, Pu X. Post-Synthetic Modification Research of Salan Titanium bis-Chelates via Sonogashira Reaction. ACTA CHIMICA SINICA 2021. [DOI: 10.6023/a21060282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
21
|
When Endoplasmic Reticulum Proteostasis Meets the DNA Damage Response. Trends Cell Biol 2020; 30:881-891. [PMID: 33036871 DOI: 10.1016/j.tcb.2020.09.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
Abstract
Sustaining both proteome and genome integrity (GI) requires the integration of a wide range of mechanisms and signaling pathways. These comprise, in particular, the unfolded protein response (UPR) and the DNA damage response (DDR). These adaptive mechanisms take place respectively in the endoplasmic reticulum (ER) and in the nucleus. UPR and DDR alterations are associated with aging and with pathologies such as degenerative diseases, metabolic and inflammatory disorders, and cancer. We discuss the emerging signaling crosstalk between UPR stress sensors and the DDR, as well as their involvement in cancer biology.
Collapse
|