1
|
Trommenschlager A, Mabrouk N, Racoeur C, Godard A, Balan C, Dubrez L, Bettaïeb A, Husson J, Le Gendre P, Paul C, Bodio E. From the Design of Innovative Ti-Pt Heterometallic Complexes to the Development of Highly Anti-Proliferative Water-Soluble Cationic Titanocenes. Chembiochem 2024; 25:e202400099. [PMID: 38749920 DOI: 10.1002/cbic.202400099] [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: 01/31/2024] [Revised: 04/19/2024] [Indexed: 07/27/2024]
Abstract
Two innovative early/late Ti-Pt-heterobimetallic complexes were synthesized, characterized, and screened in cell-based assays using several human (SW480 and MDA-MB-231) and murine cancer cell lines (CT26 and EMT6) as well as a non-cancerous cell line (HMEC). The combination of the two metals - titanium(IV) and platinum (IV) - in a single molecule led to a synergistic biological activity (higher anti-proliferative properties than a mixture of each of the corresponding monometallic complexes). This study also investigated the benefits of associating a metal-free terpyridine moiety (with intrinsic biological activity) with a water-soluble titanocene fragment. The present work reveals that these combinations results in water-soluble titanocene compounds displaying an anti-proliferative activity down to the submicromolar level. One of these complexes induced an antitumor effect in vivo in CT26 tumor bearing BALB/C mice. The terpyridine moiety was also used to track the complex in vitro by multiphoton microscopy imaging.
Collapse
Affiliation(s)
- Audrey Trommenschlager
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB UMR CNRS 6302), Université de Bourgogne, 21000, Dijon, France
| | - Nesrine Mabrouk
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC, EA7269) EPHE, PSL Research University, 75000, Paris, France, Université de Bourgogne, 21000, Dijon, France
| | - Cindy Racoeur
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC, EA7269) EPHE, PSL Research University, 75000, Paris, France, Université de Bourgogne, 21000, Dijon, France
| | - Amélie Godard
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB UMR CNRS 6302), Université de Bourgogne, 21000, Dijon, France
| | - Cédric Balan
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB UMR CNRS 6302), Université de Bourgogne, 21000, Dijon, France
| | - Laurence Dubrez
- LNC UMR1231, Institut National de la Santé et de la Recherche Médicale (Inserm), Université de Bourgogne, 21000, Dijon, France
| | - Ali Bettaïeb
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC, EA7269) EPHE, PSL Research University, 75000, Paris, France, Université de Bourgogne, 21000, Dijon, France
| | - Jérôme Husson
- Institut UTINAM UMR6213, Univ. de Franche-Comté, 25030, Besançon Cedex, France
| | - Pierre Le Gendre
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB UMR CNRS 6302), Université de Bourgogne, 21000, Dijon, France
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC, EA7269) EPHE, PSL Research University, 75000, Paris, France, Université de Bourgogne, 21000, Dijon, France
| | - Ewen Bodio
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB UMR CNRS 6302), Université de Bourgogne, 21000, Dijon, France
| |
Collapse
|
2
|
Atrián-Blasco E, Sáez J, Rodriguez-Yoldi MJ, Cerrada E. Heteronuclear Complexes with Promising Anticancer Activity against Colon Cancer. Biomedicines 2024; 12:1763. [PMID: 39200227 PMCID: PMC11351612 DOI: 10.3390/biomedicines12081763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/02/2024] Open
Abstract
This study investigates the activity of novel gold(I) and copper(I)/zinc(II) heteronuclear complexes against colon cancer. The synthesised heteronuclear Au(I)-Cu(I) and Au(I)-Zn(II) complexes were characterised and evaluated for their anticancer activity using human colon cancer cell lines (Caco-2). The complexes exhibited potent cytotoxicity, with IC50 values in the low micromolar range, and effectively induced apoptosis in cancer cells. In the case of complex [Cu{Au(Spy)(PTA)}2]PF6 (2), its cytotoxicity is ×10 higher than its mononuclear precursor, while showing low cytotoxicity towards differentiated healthy cells. Mechanistic studies revealed that complex 2 inhibits the activity of thioredoxin reductase, a key enzyme involved in redox regulation, leading to an increase in reactive oxygen species (ROS) levels and oxidative stress, in addition to an alteration in DNA's tertiary structure. Furthermore, the complexes demonstrated a strong binding affinity to bovine serum albumin (BSA), suggesting the potential for effective drug delivery and bioavailability. Collectively, these findings highlight the potential of the investigated heteronuclear Au(I)-Cu(I) and Au(I)-Zn(II) complexes as promising anticancer agents, particularly against colon cancer, through their ability to disrupt redox homeostasis and induce oxidative stress-mediated cell death.
Collapse
Affiliation(s)
- Elena Atrián-Blasco
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea—ISQCH, Consejo Superior de Investigaciones Científicas, Universidad de Zaragoza, 50009 Zaragoza, Spain (J.S.)
| | - Javier Sáez
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea—ISQCH, Consejo Superior de Investigaciones Científicas, Universidad de Zaragoza, 50009 Zaragoza, Spain (J.S.)
| | - Maria Jesús Rodriguez-Yoldi
- Departamento de Farmacología y Fisiología, Medicina Legal y Forense, Unidad de Fisiología, Facultad de Veterinaria, Ciber de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto Agroalimentario de Aragón (IA2), 50013 Zaragoza, Spain;
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Elena Cerrada
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea—ISQCH, Consejo Superior de Investigaciones Científicas, Universidad de Zaragoza, 50009 Zaragoza, Spain (J.S.)
| |
Collapse
|
3
|
Islam MT, Jang NH, Lee HJ. Natural Products as Regulators against Matrix Metalloproteinases for the Treatment of Cancer. Biomedicines 2024; 12:794. [PMID: 38672151 PMCID: PMC11048580 DOI: 10.3390/biomedicines12040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Cancers are currently the major cause of mortality in the world. According to previous studies, matrix metalloproteinases (MMPs) have an impact on tumor cell proliferation, which could lead to the onset and progression of cancers. Therefore, regulating the expression and activity of MMPs, especially MMP-2 and MMP-9, could be a promising strategy to reduce the risk of cancers. Various studies have tried to investigate and understand the pathophysiology of cancers to suggest potent treatments. In this review, we summarize how natural products from marine organisms and plants, as regulators of MMP-2 and MMP-9 expression and enzymatic activity, can operate as potent anticancer agents.
Collapse
Affiliation(s)
- Md. Towhedul Islam
- Department of Chemistry, Faculty of Science, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
| | - Nak Han Jang
- Department of Chemistry Education, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry Education, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
4
|
Xiong L, Zhang Y, Wang J, Yu M, Huang L, Hou Y, Li G, Wang L, Li Y. Novel small molecule inhibitors targeting renal cell carcinoma: Status, challenges, future directions. Eur J Med Chem 2024; 267:116158. [PMID: 38278080 DOI: 10.1016/j.ejmech.2024.116158] [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: 11/07/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024]
Abstract
Renal cell carcinoma (RCC) is the most common renal malignancy with a rapidly increasing morbidity and mortality rate gradually. RCC has a high mortality rate and an extremely poor prognosis. Despite numerous treatment strategies, RCC is resistant to conventional radiotherapy and chemotherapy. In addition, the limited clinical efficacy and inevitable resistance of multiple agents suggest an unmet clinical need. Therefore, there is an urgent need to develop novel anti-RCC candidates. Nowadays many promising results have been achieved with the development of novel small molecule inhibitors against RCC. This paper reviews the recent research progress of novel small molecule inhibitors targeting RCC. It is focusing on the structural optimization process and conformational relationships of small molecule inhibitors, as well as the potential mechanisms and anticancer activities for the treatment of RCC. To provide a theoretical basis for promoting the clinical translation of novel small molecule inhibitors, we discussed their application prospects and future development directions. It could be capable of improving the clinical efficacy of RCC and improving the therapy resistance for RCC.
Collapse
Affiliation(s)
- Lin Xiong
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Ya Zhang
- College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Min Yu
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Liming Huang
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Yanpei Hou
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Guisen Li
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Li Wang
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Yi Li
- Department of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Center for Kidney Diseases, Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
5
|
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
|
6
|
Ghardashpour M, Saeedi M, Negarandeh R, Enderami SE, Ghorbani A, Lotfizadeh A, Jafari A, Arezoumandi A, Hassannia H, Molania T. Anti-inflammatory and tissue repair effect of cinnamaldehyde and nano cinnamaldehyde on gingival fibroblasts and macrophages. BMC Oral Health 2023; 23:1014. [PMID: 38110929 PMCID: PMC10729471 DOI: 10.1186/s12903-023-03682-9] [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: 08/11/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Recurrent aphthous stomatitis has a complex and inflammatory origin. Among the great variety of medications it is increasingly common to use herbal medicines due to the adverse side effects of chemical medications. Considering the anti-inflammatory properties of cinnamaldehyde and the lack of studies related to the effectiveness of its nano form; This study investigates the effect of cinnamaldehyde and nano cinnamaldehyde on the healing rate of recurrent aphthous stomatitis lesions. METHODS In a laboratory experiment, cinnamaldehyde was converted into niosomal nanoparticles. The niosome vesicles diameter and polydispersity index were measured at 25°C using a dynamic light scattering (DLS) Mastersizer 2000 (Malvern Panalytical technologies: UK) and Zetasizer Nano ZS system (Malvern Instruments Worcestershire: UK). After characterizing these particles, the (2,3-Bis-(2-Methoxy-4-Nitro-5-Sulfophenyl)-2H-Tetrazolium-5-Carboxanilide) [XTT] assay was used to assess the toxicity of cinnamaldehyde and nano cinnamaldehyde on gingival fibroblast (HGF) and macrophage (THP-1) cells. By determining the release of TNF-α, IL-6, and TGF-β cytokines using ELISA kits, the level of tissue repair and anti-inflammatory capabilities of these two substances were evaluated. RESULTS The size and loading rate of the cinnamaldehyde nanoparticles were established after its creation. The optimized nanovesicle exhibited the following characteristics: particle size of 228.75 ± 2.38 nm, PDI of 0.244 ± 0.01, the zeta potential of -10.87 ± 1.09 mV and the drug encapsulation percentage of 66.72 ± 3.93%. PDIs range was between 0.242-0.274. The zeta potential values at 25°C were from -2.67 to -12.9 mV. The results of the XTT test demonstrated that nano cinnamaldehyde exhibited dose-dependent toxicity effects. Moreover, nano cinnamaldehyde released more TGF-β and had better reparative effects when taken at lower concentrations than cinnamaldehyde. CONCLUSION Nano cinnamaldehyde and cinnamaldehyde are effective in repairing tissue when used in non-toxic amounts. After confirmation in animal models, it is envisaged that these substances can be utilized to treat recurrent aphthous stomatitis.
Collapse
Affiliation(s)
| | - Majid Saeedi
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Negarandeh
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Ehsan Enderami
- Immunogenetics Research Center, Department of Medical Biotechnology, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anahita Ghorbani
- Department of Oral and Maxillofacial Medicine, Dental Research Center, Faculty of Dentistry, Mazandaran University of Medical Sciences, Sari, Iran
| | | | | | | | - Hadi Hassannia
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Tahereh Molania
- Department of Oral and Maxillofacial Medicine, Dental Research Center, Faculty of Dentistry, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
7
|
Ahad A, K. Saeed H, del Solar V, López-Hernández JE, Michel A, Mathew J, Lewis JS, Contel M. Shifting the Antibody-Drug Conjugate Paradigm: A Trastuzumab-Gold-Based Conjugate Demonstrates High Efficacy against Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer Mouse Model. ACS Pharmacol Transl Sci 2023; 6:1972-1986. [PMID: 38093840 PMCID: PMC10714425 DOI: 10.1021/acsptsci.3c00270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 01/17/2024]
Abstract
Antibody-drug conjugates (ADCs) combine the selectivity of monoclonal antibodies (mAbs) with the efficacy of chemotherapeutics to target cancers without toxicity to normal tissue. Clinically, most chemotherapeutic ADCs are based on complex organic molecules, while the conjugation of metallodrugs to mAbs has been overlooked, despite the resurgent interest in metal-based drugs as cancer chemotherapeutics. In 2019, we described the first gold ADCs containing gold-triphenylphosphane fragments as a proof of concept. The ADCs (based on the antibody trastuzumab) were selective and highly active against HER2-positive breast cancer cells. In this study, we developed site-specific ADCs (Thio-1b and Thio-2b) using the cysteine-engineered trastuzumab derivative THIOMAB antibody technology with gold(I)-containing phosphanes and a maleimide-based linker amenable to bioconjugation (1b and 2b). In addition, we developed lysine-directed ADCs with gold payloads based on phosphanes and N-heterocyclic carbenes featuring an activated ester moiety (2c and 5c) with trastuzumab (Tras-2c and Tras-5c) and another anti-HER2 antibody, pertuzumab (Per-2c and Per-5c). Both sets of ADCs demonstrated significant anticancer potency in vitro assays. Based on these results, one ADC (Tras-2c), containing the [Au(PEt3)] fragment present in FDA-approved auranofin, was selected for an in vivo antitumor efficacy study. Immunocompromised mice xenografted with the HER2-positive human cancer cell line SKBR-3 exhibited almost complete tumor reduction and low toxicity with intravenous administration of Tras-2c. With this highly selective targeting system, we demonstrated that a subnanomolar cytotoxicity profile in cells is not required for an impressive antitumor effect in a mouse xenograft model.
Collapse
Affiliation(s)
- Afruja Ahad
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Hiwa K. Saeed
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Virginia del Solar
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Javier E. López-Hernández
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| | - Alexa Michel
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Joshua Mathew
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Molecular
Pharmacology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Radiochemistry
and Molecular Imaging Probes Core, Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Maria Contel
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Chemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| |
Collapse
|
8
|
Rodríguez-Arce E, Gavrilov E, Alvite X, Nayeem N, León IE, Neary MC, Otero L, Gambino D, Olea Azar C, Contel M. 5-Nitrofuryl-Containing Thiosemicarbazone Gold(I) Compounds: Synthesis, Stability Studies, and Anticancer Activity. Chempluschem 2023; 88:e202300115. [PMID: 37191319 PMCID: PMC10651801 DOI: 10.1002/cplu.202300115] [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: 03/01/2023] [Revised: 04/21/2023] [Indexed: 05/17/2023]
Abstract
This work describes the synthesis of four gold(I) [AuClL] compounds containing chloro and biologically active protonated thiosemicarbazones based on 5-nitrofuryl (L=HSTC). The stability of the compounds in dichloromethane, DMSO, and DMSO/culture media solutions was investigated by spectroscopy, cyclic voltammetry, and conductimetry, indicating the formation overtime of cationic monometallic [Au(HTSC)(DMSO)]± or [Au(HTSC)2 ]± , and/or dimeric species. Neutral [{Au(TSC)}2 ] species were obtained from one of the compounds in dichlomethane/n-hexane solution and characterized by X-ray crystallography revealing a Au-Au bond, and deprotonated thiosemicarbazone (TSC). The cytotoxicity of the gold compounds and thiosemicarbazone ligands was evaluated against selected cancer cell lines and compared to that of Auranofin. Studies of the most stable, cytotoxic, and selective compound on a renal cancer cell line (Caki-1) demonstrated its relevant antimigratory and anti-angiogenic properties, and preferential accumulation in the cell nuclei. Its mode of action seems to involve interaction with DNA, and subsequent cell death via apoptosis.
Collapse
Affiliation(s)
- Esteban Rodríguez-Arce
- Departamento de Química Inorgánica y Analítica, Universidad de Chile, Casilla 233, Santiago, Chile
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Gral. Flores 2124, 11800, Montevideo, Uruguay
| | - Eric Gavrilov
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Ximena Alvite
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Gral. Flores 2124, 11800, Montevideo, Uruguay
| | - Nazia Nayeem
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Biochemistry, and Chemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - Ignacio E León
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- CEQUINOR (CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, 1900, Argentina
| | - Michelle C Neary
- Chemistry Department, Hunter College, The City University of New York, New York, NY, 10065, USA
| | - Lucía Otero
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Gral. Flores 2124, 11800, Montevideo, Uruguay
| | - Dinorah Gambino
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Gral. Flores 2124, 11800, Montevideo, Uruguay
| | - Claudio Olea Azar
- Departamento de Química Inorgánica y Analítica, Universidad de Chile, Casilla 233, Santiago, Chile
| | - María Contel
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Biochemistry, and Chemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| |
Collapse
|
9
|
López-Hernández JE, Nayeem N, Cerón-Carrasco JP, Ahad A, Hafeez A, León IE, Contel M. Platinum(IV)-Gold(I) Agents with Promising Anticancer Activity: Selected Studies in 2D and 3D Triple-Negative Breast Cancer Models. Chemistry 2023; 29:e202302045. [PMID: 37507346 PMCID: PMC10615877 DOI: 10.1002/chem.202302045] [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: 06/27/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 07/30/2023]
Abstract
New heterometallic binuclear and trinuclear platinum(IV)-gold(I) compounds of the type [Pt(L)n Cl2 (OH){(OOC-4-C6 H4 -PPh2 )AuCl}x ] (L=NH3 , n=2; x=1, 2; L=diaminocyclohexane, DACH, n=1; x=2) are described. These compounds are cytotoxic and selective against a small panel of renal, bladder, ovarian, and breast cancer cell lines. We selected a trinuclear PtAu2 compound containing the PtIV core based on oxaliplatin, to further investigate its cell-death pathway, cell and organelle uptake and anticancer effects against the triple-negative breast cancer (TNBC) MDA-MB-231 cell line. This compound induces apoptosis and accumulates mainly in the nucleus and mitochondria. It also exerts remarkable antimigratory and antiangiogenic properties, and has a potent cytotoxic effect against TNBC 3D spheroids. Trinuclear compounds do not seem to display relevant interactions with calf thymus (CT) DNA and plasmid (pBR322) even in the presence of reducing agents, but inhibit pro-angiogenic enzyme thioredoxin reductase (TrxR) in TNBC cells.
Collapse
Affiliation(s)
- Javier E López-Hernández
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - Nazia Nayeem
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - José P Cerón-Carrasco
- Centro Universitario de la Defensa, Universidad Politécnica de Cartagena, C/Coronel López Peña s/n, Base Aérea de San Javier, Santiago de la Ribera, 30720, Murcia, Spain
| | - Afruja Ahad
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
- Radiology, Molecular Pharmacology Program, and, Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, NY 11065, USA
| | - Aiman Hafeez
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Ignacio E León
- Centro de Química Inorgánica, CEQUINOR (CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N°1465, La Plata, 1900, Argentina
| | - Maria Contel
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| |
Collapse
|
10
|
Kshetri M, Jogadi W, Alqarni S, Datta P, Cheline M, Sharma A, Betters T, Broyles D, Zheng YR. Exploring the Impact of Head Group Modifications on the Anticancer Activities of Fatty-Acid-like Platinum(IV) Prodrugs: A Structure-Activity Relationship Study. Int J Mol Sci 2023; 24:13301. [PMID: 37686109 PMCID: PMC10487970 DOI: 10.3390/ijms241713301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
We conducted the first comprehensive investigation on the impact of head group modifications on the anticancer activities of fatty-acid-like Pt(IV) prodrugs (FALPs), which are a class of platinum-based metallodrugs that target mitochondria. We created a small library of FALPs (1-9) with diverse head group modifications. The outcomes of our study demonstrate that hydrophilic modifications exclusively enhance the potency of these metallodrugs, whereas hydrophobic modifications significantly decrease their cytotoxicity. To further understand this interesting structure-activity relationship, we chose two representative FALPs (compounds 2 and 7) as model compounds: one (2) with a hydrophilic polyethylene glycol (PEG) head group, and the other (7) with a hydrophobic hydrocarbon modification of the same molecular weight. Using these FALPs, we conducted a targeted investigation on the mechanism of action. Our study revealed that compound 2, with hydrophilic modifications, exhibited remarkable penetration into cancer cells and mitochondria, leading to subsequent mitochondrial and DNA damage, and effectively eradicating cancer cells. In contrast, compound 7, with hydrophobic modifications, displayed a significantly lower uptake and weaker cellular responses. The collective results present a different perspective, indicating that increased hydrophobicity may not necessarily enhance cellular uptake as is conventionally believed. These findings provide valuable new insights into the fundamental principles of developing metallodrugs.
Collapse
Affiliation(s)
- Man Kshetri
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Wjdan Jogadi
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Suha Alqarni
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
- Department of Chemistry, University of Bisha, Bisha 67714, Saudi Arabia
| | - Payel Datta
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - May Cheline
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Arpit Sharma
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Tyler Betters
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Deonya Broyles
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| | - Yao-Rong Zheng
- Department of Chemistry and Biochemistry, Kent State University, 236 Integrated Sciences Building, Kent, OH 44242, USA (S.A.); (P.D.); (M.C.)
| |
Collapse
|
11
|
López-Hernández JE, Contel M. Promising heterometallic compounds as anticancer agents: Recent studies in vivo. Curr Opin Chem Biol 2023; 72:102250. [PMID: 36566618 PMCID: PMC10880551 DOI: 10.1016/j.cbpa.2022.102250] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
Over the past decade, interest on multitarget anticancer drugs -including heterometallic compounds-has increased considerably. Heterometallic species display improved efficacy and physicochemical properties compared to the individual metallic fragments for a variety of metal pair combinations. By 2018, several compounds had emerged as promising candidates against cisplatin resistant cancers. Here, we summarize research contributions to this topic over the past four years (July 2018-July 2022). In particular, we highlight five articles reporting on the in vivo activity and preliminary mechanisms of action for five groups of compounds. From this selection, we further feature two families of compounds based on Pt(IV)-Gd(III) and Ti(IV)-Au(I) metal combinations, given their potential for clinical translation.
Collapse
Affiliation(s)
- Javier E López-Hernández
- Department of Chemistry, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY, 11210, USA; Brooklyn College Cancer Center BCCC-CURE, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY, 11210, USA; Biochemistry, The Graduate Center, The City University of New York, 365 5th Avenue, New York, 11006, USA
| | - Maria Contel
- Department of Chemistry, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY, 11210, USA; Brooklyn College Cancer Center BCCC-CURE, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, NY, 11210, USA; Biochemistry, The Graduate Center, The City University of New York, 365 5th Avenue, New York, 11006, USA; Chemistry, The Graduate Center, The City University of New York, 365 5th Avenue, New York, 11006, USA; Biology PhD Programs, The Graduate Center, The City University of New York, 365 5th Avenue, New York, 11006, USA.
| |
Collapse
|
12
|
Nguyen YT, Kim N, Lee HJ. Metal Complexes as Promising Matrix Metalloproteinases Regulators. Int J Mol Sci 2023; 24:ijms24021258. [PMID: 36674771 PMCID: PMC9861486 DOI: 10.3390/ijms24021258] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Nowadays, cancers and dementia, such as Alzheimer's disease, are the most fatal causes of death. Many studies tried to understand the pathogenesis of those diseases clearly and develop a promising way to treat the diseases. Matrix metalloproteinases (MMPs) have been reported to be involved in the pathology of cancers and AD through tumor cell movement and amyloid degradation. Therefore, control of the levels and actions of MMPs, especially MMP-2 and MMP-9, is necessary to care for and/or cure cancer and AD. Various molecules have been examined for their potential application as regulators of MMPs expression and activity. Among the molecules, multiple metal complexes have shown advantages, including simple synthesis, less toxicity and specificity toward MMPs in cancer cells or in the brain. In this review, we summarize the recent studies and knowledge of metal complexes (e.g., Pt-, Ru-, Au-, Fe-, Cu-, Ni-, Zn-, and Sn-complexes) targeting MMPs and their potentials for treating and/or caring the most fatal human diseases, cancers and AD.
Collapse
Affiliation(s)
- Yen Thi Nguyen
- Department of Chemistry, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
| | - Namdoo Kim
- Department of Chemistry, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
- Correspondence: (N.K.); (H.J.L.)
| | - Hyuck Jin Lee
- Department of Chemistry Education, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
- Correspondence: (N.K.); (H.J.L.)
| |
Collapse
|
13
|
Highlights of New Strategies to Increase the Efficacy of Transition Metal Complexes for Cancer Treatments. Molecules 2022; 28:molecules28010273. [PMID: 36615466 PMCID: PMC9822110 DOI: 10.3390/molecules28010273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
Although important progress has been made, cancer still remains a complex disease to treat. Serious side effects, the insurgence of resistance and poor selectivity are some of the problems associated with the classical metal-based anti-cancer therapies currently in clinical use. New treatment approaches are still needed to increase cancer patient survival without cancer recurrence. Herein, we reviewed two promising-at least in our opinion-new strategies to increase the efficacy of transition metal-based complexes. First, we considered the possibility of assembling two biologically active fragments containing different metal centres into the same molecule, thus obtaining a heterobimetallic complex. A critical comparison with the monometallic counterparts was done. The reviewed literature has been divided into two groups: the case of platinum; the case of gold. Secondly, the conjugation of metal-based complexes to a targeting moiety was discussed. Particularly, we highlighted some interesting examples of compounds targeting cancer cell organelles according to a third-order targeting approach, and complexes targeting the whole cancer cell, according to a second-order targeting strategy.
Collapse
|
14
|
Acharya N, Singh KP. Recent advances in the molecular basis of chemotherapy resistance and potential application of epigenetic therapeutics in chemorefractory renal cell carcinoma. WIREs Mech Dis 2022; 14:e1575. [DOI: 10.1002/wsbm.1575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/11/2022] [Accepted: 06/22/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Narayan Acharya
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH) Texas Tech University Lubbock Texas USA
| | - Kamaleshwar P. Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH) Texas Tech University Lubbock Texas USA
| |
Collapse
|
15
|
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: 54] [Impact Index Per Article: 27.0] [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
|
16
|
Banerjee S, Banerjee S. Metal-Based Complexes as Potential Anti-cancer Agents. Anticancer Agents Med Chem 2022; 22:2684-2707. [PMID: 35362388 DOI: 10.2174/1871520622666220331085144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/16/2021] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
Abstract
Metal based therapy is no new in biomedical research. In early days the biggest limitation was the inequality among therapeutical and toxicological dosages. Ever since, Barnett Rosenberg discovered cisplatin, a new era has begun to treat cancer with metal complexes. Platinum complexes such as oxaliplatin, cisplatin, and carboplatin, seem to be the foundation of metal/s-based components to challenge malignancies. With an advancement in the biomolemoecular mechanism, researchers have started developing non-classical platinum-based complexes, where a different mechanistic approach of the complexes is observed towards the biomolecular target. Till date, larger number of metal/s-based complexes was synthesized by overhauling the present structures chemically by substituting the ligand or preparing the whole novel component with improved cytotoxic and safety profiles. Howsoever, due to elevated accentuation upon the therapeutic importance of metal/s-based components, a couple of those agents are at present on clinical trials and several other are in anticipating regulatory endorsement to enter the trial. This literature highlights the detailed heterometallic multinuclear components, primarily focusing on platinum, ruthenium, gold and remarks on possible stability, synergism, mechanistic studies and structure activity relationships.
Collapse
Affiliation(s)
- Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Ashram More, G.T. Road, Asansol-713301, West Bengal, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Ashram More, G.T. Road, Asansol-713301, West Bengal, India
| |
Collapse
|
17
|
Xiao Q, Liu Y, Jiang G, Liu Y, Huang Y, Liu W, Zhang Z. Heteroleptic Gold(I)-bisNHC complex with excellent activity in vitro, ex vivo and in vivo against endometrial cancer. Eur J Med Chem 2022; 236:114302. [DOI: 10.1016/j.ejmech.2022.114302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 01/02/2023]
|
18
|
González-Ballesteros MM, Mejía C, Ruiz-Azuara L. Metallodrugs, an approach against invasion and metastasis in cancer treatment. FEBS Open Bio 2022; 12:880-899. [PMID: 35170871 PMCID: PMC9063434 DOI: 10.1002/2211-5463.13381] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/21/2022] [Accepted: 02/15/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer is a heterogeneous and multifactorial disease that causes high mortality throughout the world; therefore, finding the most effective therapies is a major research challenge. Currently, most anticancer drugs present a limited number of well‐established targets, such as cell proliferation or death; however, it is important to consider that the worse progression of cancer toward pathological stages implies invasion and metastasis processes. Medicinal Inorganic Chemistry (MIC) is a young area that deals with the design, synthesis, characterization, preclinical evaluation, and mechanism of action of new inorganic compounds, called metallodrugs. The properties of metallic ions allow enriching of strategies for the design of new drugs, enabling the adjustment of physicochemical and stereochemical properties. Metallodrugs can adopt geometries, such as tetrahedral, octahedral, square planar, and square planar pyramid, which adjusts their arrangement and facilitates binding with a wide variety of targets. The redox properties of some metal ions can be modulated by the presence of the bound ligands to adjust their interaction, thereby opening a range of mechanisms of action. In this regard, the mechanisms of action that trigger the biological activity of metallodrugs have been generally identified by: (a) coordination of the metal to biomolecules (for instance, cisplatin binds to the N7 in DNA guanine, as Pt‐N via coordination of the inhibition of enzymes); (b) redox‐active; and (c) ROS production. For this reason, a series of metallodrugs can interact with several specific targets in the anti‐invasive processes of cancer and can prevent metastasis. The structural base of several metal compounds shows great anticancer potential by inhibiting the signaling pathways related to cancer progression. In this minireview, we present the advances in the field of antimetastatic effects of metallodrugs.
Collapse
Affiliation(s)
- Mauricio M González-Ballesteros
- Departamento de Química Inorgánica y Nuclear, Facultad de Química, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Ciudad de México, CP, 04510, Mexico
| | - Carmen Mejía
- Laboratorio de Biología Celular, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Querétaro, C.P, 76230, México
| | - Lena Ruiz-Azuara
- Departamento de Química Inorgánica y Nuclear, Facultad de Química, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Ciudad de México, CP, 04510, Mexico
| |
Collapse
|
19
|
Exploring Titanium(IV) Complexes as Potential Antimicrobial Compounds. Antibiotics (Basel) 2022; 11:antibiotics11020158. [PMID: 35203761 PMCID: PMC8868518 DOI: 10.3390/antibiotics11020158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 01/27/2023] Open
Abstract
Due to the rapid mutation of pathogenic microorganisms, drug-resistant superbugs have evolved. Antimicrobial-resistant germs may share their resistance genes with other germs, making them untreatable. The search for more combative antibiotic compounds has led researchers to explore metal-based strategies centered on perturbing the bioavailability of essential metals in microbes and examining the therapeutic potential of metal complexes. Given the limited knowledge on the application of titanium(IV), in this work, eight Ti(IV) complexes and some of their corresponding ligands were screened by the Community for Open Antimicrobial Drug Discovery for antimicrobial activity. The compounds were selected for evaluation because of their low cytotoxic/antiproliferative behavior against a human non-cancer cell line. At pH 7.4, these compounds vary in terms of their solution stability and ligand exchange lability; therefore, an assessment of their solution behavior provides some insight regarding the importance of the identity of the metal compound to the antimicrobial therapeutic potential. Only one compound, Ti(deferasirox)2, exhibited promising inhibitory activity against the Gram-positive bacteria methicillin-resistant Staphylococcus aureus and minimal toxicity against human cells. The ability of this compound to undergo transmetalation with labile Fe(III) sources and, as a consequence, inhibit Fe bioavailability and ribonucleotide reductase is evaluated as a possible mechanism for its antibiotic effect.
Collapse
|
20
|
Ngoepe MP, Clayton HS. Metal Complexes as DNA Synthesis and/or Repair Inhibitors: Anticancer and Antimicrobial Agents. PHARMACEUTICAL FRONTS 2021. [DOI: 10.1055/s-0041-1741035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AbstractMedicinal inorganic chemistry involving the utilization of metal-based compounds as therapeutics has become a field showing distinct promise. DNA and RNA are ideal drug targets for therapeutic intervention in the case of various diseases, such as cancer and microbial infection. Metals play a vital role in medicine, with at least 10 metals known to be essential for human life and a further 46 nonessential metals having been involved in drug therapies and diagnosis. These metal-based complexes interact with DNA in various ways, and are often delivered as prodrugs which undergo activation in vivo. Metal complexes cause DNA crosslinking, leading to the inhibition of DNA synthesis and repair. In this review, the various interactions of metal complexes with DNA nucleic acids, as well as the underlying mechanism of action, were highlighted. Furthermore, we also discussed various tools used to investigate the interaction between metal complexes and the DNA. The tools included in vitro techniques such as spectroscopy and electrophoresis, and in silico studies such as protein docking and density-functional theory that are highlighted for preclinical development.
Collapse
Affiliation(s)
| | - Hadley S. Clayton
- Department of Chemistry, University of South Africa, Pretoria, South Africa
| |
Collapse
|
21
|
Nayeem N, Yeasmin A, Cobos SN, Younes A, Hubbard K, Contel M. Investigation of the Effects and Mechanisms of Anticancer Action of a Ru(II)-Arene Iminophosphorane Compound in Triple Negative Breast Cancer Cells. ChemMedChem 2021; 16:3280-3292. [PMID: 34329530 PMCID: PMC8571052 DOI: 10.1002/cmdc.202100325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/23/2021] [Indexed: 12/22/2022]
Abstract
Triple negative breast cancer (TNBC) is one of the breast cancers with poorer prognosis and survival rates. TNBC has a disproportionally high incidence and mortality in women of African descent. We report on the evaluation of Ru-IM (1), a water-soluble organometallic ruthenium compound, in TNBC cell lines derived from patients of European (MDA-MB-231) and African (HCC-1806) ancestry (including IC50 values, cellular and organelle uptake, cell death pathways, cell cycle, effects on migration, invasion, and angiogenesis, a preliminary proteomic analysis, and an NCI 60 cell-line panel screen). 1 was previously found highly efficacious in MDA-MB-231 cells and xenografts, with little systemic toxicity and preferential accumulation in the tumor. We observe a similar profile for this compound in the two cell lines studied, which includes high cytotoxicity, apoptotic behavior and potential antimetastatic and antiangiogenic properties. Cytokine M-CSF, involved in the PI3/AKT pathway, shows protein expression inhibition with exposure to 1. We also demonstrate a p53 independent mechanism of action.
Collapse
Affiliation(s)
- Nazia Nayeem
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - Arefa Yeasmin
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Samantha N Cobos
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| | - Ali Younes
- Department of Chemistry, Hunter College, The City University of New York, 695 Park Avenue, New York, NY, 10065, USA
| | - Karen Hubbard
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
- Biology Department, The City College of New York, The City University of New York, 160 Covent Avenue, New York, NY, 10031, USA
| | - Maria Contel
- Department of Chemistry and Brooklyn College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, NY, 10016, USA
| |
Collapse
|
22
|
Mo X, Chen K, Chen Z, Chu B, Liu D, Liang Y, Xiong J, Yang Y, Cai J, Liang F. Antitumor Activities for Two Pt(II) Complexes of Tropolone and 8-Hydroxyquinoline Derivative. Inorg Chem 2021; 60:16128-16139. [PMID: 34647723 DOI: 10.1021/acs.inorgchem.1c01763] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The reactions of cis-Pt(DMSO)2Cl2 and tropolone (HL) with 8-hydroxyquinoline (HQ) or 2-methyl-8-hydroxyquinoline (HMQ) gave [Pt(Q)(L)] (1) and [Pt(MQ)(L)] (2), which present mononuclear structures with their Pt(II) ions four-coordinated in square planar geometries. Their in vitro biological properties were evaluated by MTT assay, which showed a remarkable cytotoxic activity on the cancer cell lines. 1 shows higher cytotoxic activities on tumor cells such as T24, HeLa, A549, and NCI-H460 than complex 2 and cisplatin, with IC50 values <16 μM. Among them, an IC50 value of 3.6 ± 0.63 μM was found for complex 1 against T24 cells. It presented a tuning cytotoxic activity by substitution groups on 8-hydroxyquinoline skeleton. In our case, the substitution groups of -H are much superior to -CH3 against tumor cells. It revealed that both complexes can induce cell apoptosis by decreasing the potential of a mitochondrial membrane, enhancing reactive oxygen species and increasing Ca2+ levels of T24 cells. The T24 cell cycle can be arrested at G2 and G1 phases by complexes 1 and 2, respectively, with an upregulation for P21 and P27 expression levels and a down-regulation for cyclin A, CDK1, Cdc25A, and cyclin B expression levels. Furthermore, complex 1 exhibits satisfactory in vivo antitumor activity as revealed by the tumor inhibitory rate and the tumor weight change as well as by the cute toxicity assay and renal pathological examinations, which is close to cisplatin and much better than complex 2. All of these suggest that 1 might be a potential candidate for developing into a safe and effective anticancer agent.
Collapse
Affiliation(s)
- Xiyu Mo
- 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 541004, P.R. China
- Department of Food and Chemical Engineering, Liuzhou Institute of Technology, Liuzhou 545616, P.R. China
| | - Kaiyong Chen
- 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 541004, P.R. China
| | - Zilu Chen
- 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 541004, P.R. China
| | - Bo Chu
- 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 541004, P.R. China
| | - Dongcheng Liu
- 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 541004, P.R. China
| | - Yuning Liang
- 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 541004, P.R. China
| | - Jianwen Xiong
- Department of Food and Chemical Engineering, Liuzhou Institute of Technology, Liuzhou 545616, P.R. China
| | - Yubing Yang
- Department of Food and Chemical Engineering, Liuzhou Institute of Technology, Liuzhou 545616, P.R. China
| | - JinYuan Cai
- Department of Food and Chemical Engineering, Liuzhou Institute of Technology, Liuzhou 545616, P.R. China
| | - Fupei Liang
- 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 541004, P.R. China
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, P.R. China
| |
Collapse
|
23
|
Huang Z, King AP, Lovett J, Lai B, Woods JJ, Harris HH, Wilson JJ. Photochemistry and in vitro anticancer activity of Pt(IV)Re(I) conjugates. Chem Commun (Camb) 2021; 57:11189-11192. [PMID: 34622255 DOI: 10.1039/d1cc04669a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The photophysical and photochemical properties of two Pt(IV)Re(I) conjugates were studied via both experimental and computational methods. Both conjugates exhibit modest photocytotoxicity against ovarian cancer cells. X-ray fluorescence microscopy showed that Pt and Re colocalize in cells whether they had been irradiated or not. This work demonstrates the potential of photoactivated multilimetallic agents for combating cancer.
Collapse
Affiliation(s)
- Zhouyang Huang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA.
| | - A Paden King
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA.
| | - James Lovett
- Department of Chemistry, The University of Adelaide, South Australia 5005, Australia
| | - Barry Lai
- Advanced Photon Source, X-ray Science Division, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Joshua J Woods
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA. .,Robert F. Smith School for Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Hugh H Harris
- Department of Chemistry, The University of Adelaide, South Australia 5005, Australia
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
24
|
Abstract
Gold compounds have been employed throughout history to treat various types of disease, from ancient times to the present day. In the year 1985, auranofin, a gold-containing compound, was approved by U.S. Food and Drug Administration (FDA) as a therapeutic agent to target rheumatoid arthritis that would facilitate easy oral drug administration as opposed to conventional intramuscular injection used in treatments. Furthermore, auranofin demonstrates promising results for the treatment of various diseases beyond rheumatoid arthritis, including cancer, neurodegenerative diseases, acquired immune deficiency syndrome, and bacterial and parasitic infections. Various potential novel applications for auranofin have been proposed for treating human diseases. Auranofin has previously been demonstrated to inhibit thioredoxin reductase (TrxR) involved within the thioredoxin (Trx) system that comprises one of the critical cellular redox systems within the body. TrxR comprises the sole known enzyme that catalyzes Trx reduction. With cancers in particular, TrxR inhibition facilitates an increase in cellular oxidative stress and suppresses tumor growth. In this review, we describe the potential of auranofin to serve as an anticancer agent and further drug repurposing to utilize this as a strategy for further appropriate drug developments.
Collapse
Affiliation(s)
- Isao Momose
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation
| | - Takefumi Onodera
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation
| |
Collapse
|
25
|
Steel TR, Tong KK, Söhnel T, Jamieson SM, Wright LJ, Crowley JD, Hanif M, Hartinger CG. Homodinuclear organometallics of ditopic N,N-chelates: Synthesis, reactivity and in vitro anticancer activity. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2020.120220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
26
|
Long Y, Cao B, Xiong X, Chan ASC, Sun RW, Zou T. Bioorthogonal Activation of Dual Catalytic and Anti‐Cancer Activities of Organogold(I) Complexes in Living Systems. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202013366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yan Long
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation School of Pharmaceutical Sciences Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Bei Cao
- Warshel Institute for Computational Biology General Education Division The Chinese University of Hong Kong Shenzhen 518172 P. R. China
| | - Xiaolin Xiong
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation School of Pharmaceutical Sciences Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Albert S. C. Chan
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation School of Pharmaceutical Sciences Sun Yat-Sen University Guangzhou 510006 P. R. China
| | | | - Taotao Zou
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation School of Pharmaceutical Sciences Sun Yat-Sen University Guangzhou 510006 P. R. China
- State Key Laboratory of Coordination Chemistry Nanjing University Nanjing 210093 P. R. China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources Guangxi Normal University Guilin 541004 P. R. China
| |
Collapse
|
27
|
Long Y, Cao B, Xiong X, Chan ASC, Sun RW, Zou T. Bioorthogonal Activation of Dual Catalytic and Anti‐Cancer Activities of Organogold(I) Complexes in Living Systems. Angew Chem Int Ed Engl 2020; 60:4133-4141. [DOI: 10.1002/anie.202013366] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/03/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Yan Long
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation School of Pharmaceutical Sciences Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Bei Cao
- Warshel Institute for Computational Biology General Education Division The Chinese University of Hong Kong Shenzhen 518172 P. R. China
| | - Xiaolin Xiong
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation School of Pharmaceutical Sciences Sun Yat-Sen University Guangzhou 510006 P. R. China
| | - Albert S. C. Chan
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation School of Pharmaceutical Sciences Sun Yat-Sen University Guangzhou 510006 P. R. China
| | | | - Taotao Zou
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation School of Pharmaceutical Sciences Sun Yat-Sen University Guangzhou 510006 P. R. China
- State Key Laboratory of Coordination Chemistry Nanjing University Nanjing 210093 P. R. China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources Guangxi Normal University Guilin 541004 P. R. China
| |
Collapse
|
28
|
Wang Y, Jin J, Shu L, Li T, Lu S, Subarkhan MKM, Chen C, Wang H. New Organometallic Ruthenium(II) Compounds Synergistically Show Cytotoxic, Antimetastatic and Antiangiogenic Activities for the Treatment of Metastatic Cancer. Chemistry 2020; 26:15170-15182. [PMID: 32639591 DOI: 10.1002/chem.202002970] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Indexed: 12/20/2022]
Abstract
In this study, we newly designed and synthesized a small library of ten structurally related C,N-cyclometalated ruthenium(II) complexes containing various pyridine-functionalized NHC ligand and chelating bipyridyl ligands (e.g., 2,2'-bipyridine, 5,5'-dimethyl-2,2'-bipyridine, and 1,10-phenanthroline (phen)). The complexes were well characterized by NMR, electrospray ionization-mass spectrometry, and single-crystal X-ray structure analyses. Among the new ruthenium(II) derivatives, we identified that the complex Ru8 bearing bulky moieties (i.e., phen and pentamethyl benzene) had the most potent cytotoxicity against all tested cancer cell lines, generating dose- and cell line-dependent IC50 values at the range of 3.3-15.0 μm. More significantly, Ru8 not only efficiently inhibited the metastasis process against invasion and migration of tumor cells but also exhibited potent antivascular effects by suppressing HUVEC cells migration and tube formation in vitro and blocking vessel generation in vivo (chicken chorioallantoic membrane model). In a metastatic A2780 tumor xenograft-bearing mouse model, administration of Ru8 outperformed antimetastatic agent NAMI-A and clinically approved cisplatin in terms of antitumor efficacy and inhibition of metastases to other organs. Overall, these data provided compelling evidence that the new cyclometalated ruthenium complex Ru8 is an attractive agent because of synergistically suppressing bulky tumors and metastasized tumor nudes. Therefore, the complex Ru8 deserves further investigations.
Collapse
Affiliation(s)
- Yuchen Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China.,Department of Chemical Engineering, Zhejiang University, Hangzhou, 310027, P.R. China
| | - Jiahui Jin
- Xingzhi College, Zhejiang Normal University, Jinhua, 321004, P.R. China
| | - Liwei Shu
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Tongyu Li
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Siming Lu
- Department of Laboratory Medicine, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Mohamed Kasim Mohamed Subarkhan
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| | - Chao Chen
- College of Life Sciences, Huzhou University, Huzhou, 313000, P.R. China
| | - Hangxiang Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P.R. China
| |
Collapse
|
29
|
Maciel LLF, de Freitas WR, Bull ES, Fernandes C, Horn A, de Aquino Almeida JC, Kanashiro MM. In vitro and in vivo anti-proliferative activity and ultrastructure investigations of a copper(II) complex toward human lung cancer cell NCI-H460. J Inorg Biochem 2020; 210:111166. [DOI: 10.1016/j.jinorgbio.2020.111166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 02/08/2023]
|
30
|
Elie BT, Hubbard K, Layek B, Yang WS, Prabha S, Ramos JW, Contel M. Auranofin-Based Analogues Are Effective Against Clear Cell Renal Carcinoma In Vivo and Display No Significant Systemic Toxicity. ACS Pharmacol Transl Sci 2020. [PMID: 32832867 DOI: 10.1021/acsptsci.9b00107/asset/images/large/pt9b00107_0002.jpeg] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Effective pharmacological treatments for patients with advanced clear cell renal carcinoma (ccRCC) are limited. Bimetallic titanium-gold containing compounds exhibit significant cytotoxicity against ccRCC in vitro and in vivo and inhibit invasion and angiogenisis in vitro and markers driving these phenomena. However, in vivo preclinical evaluations of such compounds have not examined their pharmacokinetics, pathology, and hematology. Here we use NOD.CB17-Prkdc SCID/J mice bearing xenograft ccRCC Caki-1 tumors to evaluate the in vivo efficacies of two titanium-gold compounds Titanocref and Titanofin (based on auranofin analogue scaffolds) accompanied by pharmacokinetic and pathology studies. A therapeutic trial was performed over 21 days at 5 mg/kg/72h of Titanocref and 10 mg/kg/72h of Titanofin tracking changes in tumor size. We observed a significant reduction of 51% and 60%, respectively (p < 0.01) in tumor size in the Titanocref- and Titanofin-treated mice compared to the starting size, while the vehicle-treated mice exhibited a tumor size increase of 138% (p < 0.01). Importantly, no signs of pathological complication as a result of treatment were found. In addition, Titanocref and Titanofin treatment reduced angiogenesis by 38% and 54%, respectively. Microarray and qRT-PCR analysis of ccRCC Caki-1 cells treated with Titanocref revealed that the compound alters apoptosis, JNK MAP kinase, and ROS pathways within 3 h of treatment. We further show activation of apoptosis by Titanocref and Titanofin in vivo by caspase 3 assay. Titanocref is active against additional kidney cancer cells. Titanocref and Titanofin are therefore promising candidates for further evaluation toward clinical application in the treatment of ccRCC.
Collapse
Affiliation(s)
- Benelita T Elie
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States
| | - Karen Hubbard
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Department of Biology, City College of New York, The City University of New York, New York, New York 10031, United States
| | - Buddhadev Layek
- University of Minnesota, College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Won Seok Yang
- University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Swayam Prabha
- University of Minnesota, College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Joe W Ramos
- University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Maria Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| |
Collapse
|
31
|
Wilson CS, Prior TJ, Sandland J, Savoie H, Boyle RW, Murray BS. Homo‐ and Hetero‐dinuclear Arene‐Linked Osmium(II) and Ruthenium(II) Organometallics: Probing the Impact of Metal Variation on Reactivity and Biological Activity. Chemistry 2020; 26:11593-11603. [DOI: 10.1002/chem.202002052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/31/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Christopher S. Wilson
- Department of Chemistry and Biochemistry University of Hull Cottingham Road Hull HU6 7RX UK
| | - Timothy J. Prior
- Department of Chemistry and Biochemistry University of Hull Cottingham Road Hull HU6 7RX UK
| | - Jordon Sandland
- Department of Chemistry and Biochemistry University of Hull Cottingham Road Hull HU6 7RX UK
| | - Huguette Savoie
- Department of Chemistry and Biochemistry University of Hull Cottingham Road Hull HU6 7RX UK
| | - Ross W. Boyle
- Department of Chemistry and Biochemistry University of Hull Cottingham Road Hull HU6 7RX UK
| | - Benjamin S. Murray
- Department of Chemistry and Biochemistry University of Hull Cottingham Road Hull HU6 7RX UK
| |
Collapse
|
32
|
Sarpong-Kumankomah S, Contel M, Gailer J. SEC hyphenated to a multielement-specific detector unravels the degradation pathway of a bimetallic anticancer complex in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1145:122093. [PMID: 32305711 PMCID: PMC7328787 DOI: 10.1016/j.jchromb.2020.122093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/13/2020] [Accepted: 03/28/2020] [Indexed: 12/14/2022]
Abstract
The bimetallic metal complex Titanocref exhibits relevant anticancer activity, but it is unknown if it is stable to reach target tissues intact. To gain insight, a pharmacologically relevant dose was added to human blood plasma and the mixture was incubated at 37 °C. The obtained mixture was analyzed 5 and 60 min later by size-exclusion chromatography hyphenated to an inductively coupled plasma atomic emission spectrometer (SEC-ICP-AES). We simultaneously detected several titanium (Ti), gold (Au) and sulfur (S)-peaks, which corresponded to a Ti degradation product that eluted partially, and a Au degradation product that eluted entirely bound to plasma proteins (both time points). Although ~70% of the intact Titanocref was retained on the column after 60 min, our results allowed us to establish - for the first time - its likely degradation pathway in human plasma at near physiological conditions. These results suggest that ~70% of Titanocref remain in plasma after 60 min, which supports results from a recent in vivo study in which mice were treated with Titanocref and revealed Ti:Au molar ratios in tumors and organs close to 1:1. Thus, our stability studies suggest that the intact drug is able to reach target tissue. Overall, our results exemplify that SEC-ICP-AES enables the execution of intermediate in vitro studies with human plasma in the context of advancing bimetallic metal-based drugs to more costly clinical studies.
Collapse
Affiliation(s)
- Sophia Sarpong-Kumankomah
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Maria Contel
- Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York 11210, USA; Chemistry, The Graduate Center, The City University of New York, 365 5th Avenue, New York 10016, USA; Biochemistry, The Graduate Center, The City University of New York, 365 5th Avenue, New York 10016, USA; Biology PhD Programs, The Graduate Center, The City University of New York, 365 5th Avenue, New York 10016, USA
| | - Jürgen Gailer
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
33
|
Tunes LG, Morato RE, Garcia A, Schmitz V, Steindel M, Corrêa-Junior JD, Dos Santos HF, Frézard F, de Almeida MV, Silva H, Moretti NS, de Barros ALB, do Monte-Neto RL. Preclinical Gold Complexes as Oral Drug Candidates to Treat Leishmaniasis Are Potent Trypanothione Reductase Inhibitors. ACS Infect Dis 2020; 6:1121-1139. [PMID: 32283915 DOI: 10.1021/acsinfecdis.9b00505] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The drugs currently used to treat leishmaniases have limitations concerning cost, efficacy, and safety, making the search for new therapeutic approaches urgent. We found that the gold(I)-derived complexes were active against L. infantum and L. braziliensis intracellular amastigotes with IC50 values ranging from 0.5 to 5.5 μM. All gold(I) complexes were potent inhibitors of trypanothione reductase (TR), with enzyme IC50 values ranging from 1 to 7.8 μM. Triethylphosphine-derived complexes enhanced reactive oxygen species (ROS) production and decreased mitochondrial respiration after 2 h of exposure, indicating that gold(I) complexes cause oxidative stress by direct ROS production, by causing mitochondrial damage or by impairing TR activity and thus accumulating ROS. There was no cross-resistance to antimony; in fact, SbR (antimony-resistant mutants) strains were hypersensitive to some of the complexes. BALB/c mice infected with luciferase-expressing L. braziliensis or L. amazonensis and treated orally with 12.5 mg/kg/day of AdT Et (3) or AdO Et (4) presented reduced lesion size and parasite burden, as revealed by bioimaging. The combination of (3) and miltefosine allowed for a 50% reduction in miltefosine treatment time. Complexes 3 and 4 presented favorable pharmacokinetic and toxicity profiles that encourage further drug development studies. Gold(I) complexes are promising antileishmanial agents, with a potential for therapeutic use, including in leishmaniasis caused by antimony-resistant parasites.
Collapse
Affiliation(s)
- Luiza G. Tunes
- Instituto René Rachou/Fiocruz Minas−Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brasil
| | - Roberta E. Morato
- Instituto René Rachou/Fiocruz Minas−Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brasil
| | - Adriana Garcia
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, Brasil
| | - Vinicius Schmitz
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, Brasil
| | - Mario Steindel
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis 88040-900, Brasil
| | - José D. Corrêa-Junior
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brasil
| | - Hélio F. Dos Santos
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, Brasil
| | - Frédéric Frézard
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brasil
| | - Mauro V. de Almeida
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, Brasil
| | - Heveline Silva
- Departamento de Química, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brasil
| | - Nilmar S. Moretti
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo 04023-062, Brasil
| | - André L. B. de Barros
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brasil
| | | |
Collapse
|
34
|
Elie BT, Hubbard K, Layek B, Yang WS, Prabha S, Ramos JW, Contel M. Auranofin-Based Analogues Are Effective Against Clear Cell Renal Carcinoma In Vivo and Display No Significant Systemic Toxicity. ACS Pharmacol Transl Sci 2020; 3:644-654. [PMID: 32832867 DOI: 10.1021/acsptsci.9b00107] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Indexed: 01/03/2023]
Abstract
Effective pharmacological treatments for patients with advanced clear cell renal carcinoma (ccRCC) are limited. Bimetallic titanium-gold containing compounds exhibit significant cytotoxicity against ccRCC in vitro and in vivo and inhibit invasion and angiogenisis in vitro and markers driving these phenomena. However, in vivo preclinical evaluations of such compounds have not examined their pharmacokinetics, pathology, and hematology. Here we use NOD.CB17-Prkdc SCID/J mice bearing xenograft ccRCC Caki-1 tumors to evaluate the in vivo efficacies of two titanium-gold compounds Titanocref and Titanofin (based on auranofin analogue scaffolds) accompanied by pharmacokinetic and pathology studies. A therapeutic trial was performed over 21 days at 5 mg/kg/72h of Titanocref and 10 mg/kg/72h of Titanofin tracking changes in tumor size. We observed a significant reduction of 51% and 60%, respectively (p < 0.01) in tumor size in the Titanocref- and Titanofin-treated mice compared to the starting size, while the vehicle-treated mice exhibited a tumor size increase of 138% (p < 0.01). Importantly, no signs of pathological complication as a result of treatment were found. In addition, Titanocref and Titanofin treatment reduced angiogenesis by 38% and 54%, respectively. Microarray and qRT-PCR analysis of ccRCC Caki-1 cells treated with Titanocref revealed that the compound alters apoptosis, JNK MAP kinase, and ROS pathways within 3 h of treatment. We further show activation of apoptosis by Titanocref and Titanofin in vivo by caspase 3 assay. Titanocref is active against additional kidney cancer cells. Titanocref and Titanofin are therefore promising candidates for further evaluation toward clinical application in the treatment of ccRCC.
Collapse
Affiliation(s)
- Benelita T Elie
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States
| | - Karen Hubbard
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Department of Biology, City College of New York, The City University of New York, New York, New York 10031, United States
| | - Buddhadev Layek
- University of Minnesota, College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Won Seok Yang
- University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Swayam Prabha
- University of Minnesota, College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Joe W Ramos
- University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Maria Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| |
Collapse
|
35
|
Guarra F, Busto N, Guerri A, Marchetti L, Marzo T, García B, Biver T, Gabbiani C. Cytotoxic Ag(I) and Au(I) NHC-carbenes bind DNA and show TrxR inhibition. J Inorg Biochem 2020; 205:110998. [DOI: 10.1016/j.jinorgbio.2020.110998] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/18/2019] [Accepted: 01/12/2020] [Indexed: 02/07/2023]
|
36
|
Syntheses of terpyridine-pyridylbenzothiazole linked ditopic ligands and their copper(II) complexes. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
37
|
Fernandez-Vega L, Ruiz Silva VA, Domínguez-González TM, Claudio-Betancourt S, Toro-Maldonado RE, Capre Maso LC, Ortiz KS, Pérez-Verdejo JA, González JR, Rosado-Fraticelli GT, Meléndez FP, Betancourt Santiago FM, Rivera-Rivera DA, Navarro CM, Bruno Chardón AC, Vera AO, Tinoco AD. Evaluating Ligand Modifications of the Titanocene and Auranofin Moieties for the Development of More Potent Anticancer Drugs. INORGANICS 2020; 8. [PMID: 34046448 PMCID: PMC8152503 DOI: 10.3390/inorganics8020010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over time platinum-based anticancer drugs have dominated the market, but their side effects significantly impact the quality of life of patients. Alternative treatments are being developed all over the world. The titanocene and auranofin families of compounds, discovered through an empirical search for other metal-based therapeutics, hold tremendous promise to improve the outcomes of cancer treatment. Herein we present a historical perspective of these compounds and review current efforts focused on the evolution of their ligands to improve their physiological solution stability, cancer selectivity, and antiproliferative performance, guided by a clear understanding of the coordination chemistry and aqueous speciation of the metal ions, of the cytotoxic mechanism of action of the compounds, and the external factors that limit their therapeutic potential. Newer members of these families of compounds and their combination in novel bimetallic complexes are the result of years of scientific research. We believe that this review can have a positive impact in the development and understanding of the metal-based drugs of gold, titanium, and beyond.
Collapse
|
38
|
Tabrizi L, Olasunkanmi LO, Fadare OA. De novodesign of thioredoxin reductase-targeted heterometallic titanocene–gold compounds of chlorambucil for mechanistic insights into renal cancer. Chem Commun (Camb) 2020; 56:297-300. [DOI: 10.1039/c9cc07406f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A chlorambucil-alkynyl mononuclear gold(i) complex and heteronuclear titanocene–gold(i) complex were studied for mechanism of action in renal cancer by experimental and computational methods.
Collapse
Affiliation(s)
- Leila Tabrizi
- School of Chemistry
- National University of Ireland
- Galway
- Ireland
| | - Lukman O. Olasunkanmi
- Department of Chemistry
- Faculty of Science
- Obafemi Awolowo University
- Ile-Ife 220005
- Nigeria
| | - Olatomide A. Fadare
- Department of Chemistry
- Faculty of Science
- Obafemi Awolowo University
- Ile-Ife 220005
- Nigeria
| |
Collapse
|
39
|
Mármol I, Castellnou P, Alvarez R, Gimeno MC, Rodríguez-Yoldi MJ, Cerrada E. Alkynyl Gold(I) complexes derived from 3-hydroxyflavones as multi-targeted drugs against colon cancer. Eur J Med Chem 2019; 183:111661. [PMID: 31546196 DOI: 10.1016/j.ejmech.2019.111661] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/20/2019] [Accepted: 08/29/2019] [Indexed: 01/26/2023]
Abstract
The design of multi-targeted drugs has gained considerable interest in the last decade thanks to their advantages in the treatment of different diseases, including cancer. The simultaneous inhibition of selected targets from cancerous cells to induce their death represents an attractive objective for the medicinal chemist in order to enhance the efficiency of chemotherapy. In the present work, several alkynyl gold(I) phosphane complexes derived from 3-hydroxyflavones active against three human cancer cell lines, colorectal adenocarcinoma Caco-2/TC7, breast adenocarcinoma MCF-7 and hepatocellular carcinoma HepG2, have been synthesized and characterized. Moreover, these compounds display high selective index values towards differentiated Caco-2 cells, which are considered as a model of non-cancerous cells. The antiproliferative effect of the most active complexes [Au(L2b)PPh3] (3b) and [Au(L2c)PTA] (4c) on Caco-2 cells, seems to be mediated by the inhibition of the enzyme cyclooxygenase-1/2 and alteration of the activities of the redox enzymes thioredoxin reductase and glutathione reductase. Both complexes triggered cell death by apoptosis, alterations in cell cycle progression and increased of ROS production. These results provide support for the suggestion that multi-targeting approach involving the interaction with cyclooxygenase-1/2 and the redox enzymes that increases ROS production, enhances cell death in vitro. All these results indicate that complexes [Au(L2b)PPh3] and [Au(L2c)PTA] are promising antiproliferative agents for further anticancer drug development.
Collapse
Affiliation(s)
- Inés Mármol
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., 50009, Zaragoza, Spain; Departamento de Farmacología y Fisiología, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013, Zaragoza, Spain
| | - Pilar Castellnou
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., 50009, Zaragoza, Spain
| | - Raquel Alvarez
- Departamento de Farmacología y Fisiología, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013, Zaragoza, Spain
| | - M Concepción Gimeno
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., 50009, Zaragoza, Spain
| | - M Jesús Rodríguez-Yoldi
- Departamento de Farmacología y Fisiología, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013, Zaragoza, Spain.
| | - Elena Cerrada
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., 50009, Zaragoza, Spain.
| |
Collapse
|
40
|
Elie BT, Hubbard K, Pechenyy Y, Layek B, Prabha S, Contel M. Preclinical evaluation of an unconventional ruthenium-gold-based chemotherapeutic: RANCE-1, in clear cell renal cell carcinoma. Cancer Med 2019; 8:4304-4314. [PMID: 31192543 PMCID: PMC6675714 DOI: 10.1002/cam4.2322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND There are few effective treatments for patients with advanced clear cell renal cell carcinoma (CCRCC). Recent findings indicate that ruthenium-gold containing compounds exhibit significant antitumor efficacy against CCRCC in vitro affecting cell viability as well as angiogenesis and markers driving those 2 phenomena. However, no in vivo preclinical evaluation of this class of compounds has been reported. METHODS Following the dose-finding pharmacokinetic determination, NOD.CB17-Prkdc SCID/J mice bearing xenograft CCRCC Caki-1 tumors were treated in an intervention trial for 21 days at 10 mg/kg/72h of RANCE-1. At the end of the trial, tumor samples were analyzed for histopathological and changes in protein expression levels were assessed. RESULTS After 21 days of treatment there was no significant change in tumor size in the RANCE-1-treated mice as compared to the starting size (+3.87%) (P = 0.082) while the vehicle treated mice exhibited a significant tumor size increase (+138%) (P < 0.01). There were no signs of pathological complications as a result of treatment. Significant reduction in the expression of VEGF, PDGF, FGF, EGFR, and HGRF, all key to the proliferation of tumor cells and stromal cells serving protumorigenic purposes was observed. CONCLUSIONS The tumor growth inhibition displayed and favorable pathology profile of RANCE-1 makes it a promising candidate for further evaluation toward clinical use for the treatment of advanced CCRCC.
Collapse
Affiliation(s)
- Benelita T. Elie
- Department of ChemistryBrooklyn College, The City University of New YorkBrooklynNew York
- Biology PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
| | - Karen Hubbard
- Biology PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
- Department of BiologyCity College of New York, The City University of New YorkNew YorkNew York
| | - Yuriy Pechenyy
- Department of BiologyCity College of New York, The City University of New YorkNew YorkNew York
| | - Buddhadev Layek
- University of Minnesota College of PharmacyMinneapolisMinnesota
| | - Swayam Prabha
- University of Minnesota College of PharmacyMinneapolisMinnesota
| | - Maria Contel
- Department of ChemistryBrooklyn College, The City University of New YorkBrooklynNew York
- Biology PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
- Chemistry PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
- Biochemistry PhD Program, The Graduate CenterThe City University of New YorkNew YorkNew York
| |
Collapse
|
41
|
van Niekerk A, Chellan P, Mapolie SF. Heterometallic Multinuclear Complexes as Anti-Cancer Agents-An Overview of Recent Developments. Eur J Inorg Chem 2019. [DOI: 10.1002/ejic.201900375] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Annick van Niekerk
- Department of Chemistry and Polymer Science; Stellenbosch University; Private bag X1, Matieland 7602 Stellenbosch South Africa
| | - Prinessa Chellan
- Department of Chemistry and Polymer Science; Stellenbosch University; Private bag X1, Matieland 7602 Stellenbosch South Africa
| | - Selwyn F. Mapolie
- Department of Chemistry and Polymer Science; Stellenbosch University; Private bag X1, Matieland 7602 Stellenbosch South Africa
| |
Collapse
|
42
|
Curado N, Giménez N, Miachin K, Aliaga-Lavrijsen M, Cornejo MA, Jarzecki AA, Contel M. Preparation of Titanocene-Gold Compounds Based on Highly Active Gold(I)-N-Heterocyclic Carbene Anticancer Agents: Preliminary in vitro Studies in Renal and Prostate Cancer Cell Lines. ChemMedChem 2019; 14:1086-1095. [PMID: 30924298 PMCID: PMC7181960 DOI: 10.1002/cmdc.201800796] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/04/2019] [Indexed: 12/14/2022]
Abstract
Heterometallic titanocene-based compounds containing gold(I)-phosphane fragments have been extremely successful against renal cancer in vitro and in vivo. The exchange of phosphane by N-heterocyclic carbene ligands to improve or modulate their pharmacological profile afforded bimetallic complexes effective against prostate cancer, but less effective against renal cancer in vitro. Herein we report the synthesis of new bimetallic Ti-Au compounds by the incorporation of two previously reported highly active gold(I)-N-heterocyclic carbene fragments derived from 4,5-diarylimidazoles. The two new compounds [(η5 -C5 H5 )2 TiMe(μ-mba)Au(NHC)] (where NHC=1,3-dibenzyl-4,5-diphenylimidazol-2-ylidene, NHC-Bn 2 a; or 1,3-diethyl-4,5-diphenylimidazol-2-ylidene, NHC-Et 2 b) with the dual linker (-OC(O)-p-C6 H4 -S-) containing both a carboxylate and a thiolate group were evaluated in vitro against renal and prostate cancer cell lines. The compounds were found to be more cytotoxic than previously described Ti-Au compounds containing non-optimized gold(I)-N-heterocyclic fragments. We present studies to evaluate their effects on cell death pathways, migration, inhibition of thioredoxin reductase (TrxR) and vascular endothelial growth factor (VEGF) in the PC3 prostate cancer cell line. The results show that the incorporation of a second metallic fragment such as titanocene into biologically active gold(I) compounds improves their pharmacological profile.
Collapse
Affiliation(s)
- Natalia Curado
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Nora Giménez
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Departamento de Química, Centro de Síntesis Química de la Rioja (CISQ), Universidad de la Rioja, 2606, Logroño, Spain
| | - Kirill Miachin
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Mélanie Aliaga-Lavrijsen
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Departamento de Química Inorganica, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-Universidad de Zaragoza, 5009, Zararagoza, Spain
| | - Mike A Cornejo
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Andrzej A Jarzecki
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Chemistry, Biochemistry and Biology PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA
| | - María Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
- Chemistry, Biochemistry and Biology PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA
| |
Collapse
|
43
|
Parveen S, Hanif M, Leung E, Tong KKH, Yang A, Astin J, De Zoysa GH, Steel TR, Goodman D, Movassaghi S, Söhnel T, Sarojini V, Jamieson SMF, Hartinger CG. Anticancer organorhodium and -iridium complexes with low toxicity in vivo but high potency in vitro: DNA damage, reactive oxygen species formation, and haemolytic activity. Chem Commun (Camb) 2019; 55:12016-12019. [DOI: 10.1039/c9cc03822a] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dinuclear RhIII(Cp*) and IrIII(Cp*) complexes demonstrated potent in vitro anticancer activity while exhibiting low toxicity in haemolysis studies and in vivo zebrafish models.
Collapse
Affiliation(s)
- Shahida Parveen
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - Muhammad Hanif
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - Euphemia Leung
- Auckland Cancer Society Research Centre
- University of Auckland
- Auckland 1142
- New Zealand
| | - Kelvin K. H. Tong
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - Annie Yang
- Department of Molecular Medicine and Pathology
- University of Auckland
- Auckland 1142
- New Zealand
| | - Jonathan Astin
- Department of Molecular Medicine and Pathology
- University of Auckland
- Auckland 1142
- New Zealand
| | - Gayan H. De Zoysa
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - Tasha R. Steel
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - David Goodman
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - Sanam Movassaghi
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - Tilo Söhnel
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | | | | | | |
Collapse
|
44
|
de la Cueva-Alique I, Muñoz-Moreno L, de la Torre-Rubio E, Bajo AM, Gude L, Cuenca T, Royo E. Water soluble, optically active monofunctional Pd(ii) and Pt(ii) compounds: promising adhesive and antimigratory effects on human prostate PC-3 cancer cells. Dalton Trans 2019; 48:14279-14293. [DOI: 10.1039/c9dt02873k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Water soluble, enantiomerically pure “rule breakers” Pd(ii) and Pt(ii) compounds with promising anticancer potential are reported.
Collapse
Affiliation(s)
- Isabel de la Cueva-Alique
- Departamento de Química Orgánica y Química Inorgánica
- Instituto de Investigación en Química Andrés M. del Río (IQAR)
- Universidad de Alcalá
- Madrid
- Spain
| | - Laura Muñoz-Moreno
- Departamento de Biología de Sistemas
- Facultad de Medicina y Ciencias de la Salud
- Universidad de Alcalá
- Madrid
- Spain
| | - Elena de la Torre-Rubio
- Departamento de Química Orgánica y Química Inorgánica
- Instituto de Investigación en Química Andrés M. del Río (IQAR)
- Universidad de Alcalá
- Madrid
- Spain
| | - Ana M. Bajo
- Departamento de Biología de Sistemas
- Facultad de Medicina y Ciencias de la Salud
- Universidad de Alcalá
- Madrid
- Spain
| | - Lourdes Gude
- Departamento de Química Orgánica y Química Inorgánica
- Instituto de Investigación en Química Andrés M. del Río (IQAR)
- Universidad de Alcalá
- Madrid
- Spain
| | - Tomás Cuenca
- Departamento de Química Orgánica y Química Inorgánica
- Instituto de Investigación en Química Andrés M. del Río (IQAR)
- Universidad de Alcalá
- Madrid
- Spain
| | - Eva Royo
- Departamento de Química Orgánica y Química Inorgánica
- Instituto de Investigación en Química Andrés M. del Río (IQAR)
- Universidad de Alcalá
- Madrid
- Spain
| |
Collapse
|