1
|
Predarska I, Kaluđerović GN, Hey-Hawkins E. Nanostructured mesoporous silica carriers for platinum-based conjugates with anti-inflammatory agents. BIOMATERIALS ADVANCES 2024; 165:213998. [PMID: 39236581 DOI: 10.1016/j.bioadv.2024.213998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 08/09/2024] [Accepted: 08/17/2024] [Indexed: 09/07/2024]
Abstract
This review discusses the relationship between inflammation and cancer initiation and progression, which has prompted research into anti-inflammatory approaches for cancer prevention and treatment. Specifically, it focuses on the use of inflammation-reducing agents to enhance the effectiveness of tumor treatment methods. These agents are combined with platinum(II)-based antitumor drugs to create multifunctional platinum(IV) prodrugs, allowing for simultaneous delivery to tumor cells in a specific ratio. Once inside the cells and subjected to intracellular reduction, both components can act in parallel through distinct pathways. Motivated by the objective of reducing the systemic toxicity associated with contemporary chemotherapy, and with the aim of leveraging the passive enhanced permeability and retention effect exhibited by nanostructured materials to improve their accumulation within tumor tissues, the platinum(IV) complexes have been efficiently loaded into mesoporous silica SBA-15 material. The resulting nanostructured materials are capable of providing controlled release of the conjugates when subjected to simulated plasma conditions. This feature suggests the potential for extended circulation within the body in vivo, with minimal premature release of the drug before reaching the intended target site. The primary emphasis of this review is on research that integrates these two approaches to develop chemotherapeutic treatments that are both more efficient and less harmful.
Collapse
Affiliation(s)
- Ivana Predarska
- Leipzig University, Faculty of Chemistry and Mineralogy, Centre for Biotechnology and Biomedicine (BBZ), Institute of Bioanalytical Chemistry, Deutscher Platz 5, 04103 Leipzig, Germany; Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Goran N Kaluđerović
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany.
| | - Evamarie Hey-Hawkins
- Leipzig University, Faculty of Chemistry and Mineralogy, Centre for Biotechnology and Biomedicine (BBZ), Institute of Bioanalytical Chemistry, Deutscher Platz 5, 04103 Leipzig, Germany.
| |
Collapse
|
2
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
3
|
Liu B, Liang BB, Cao WD, Su XX, Cao Q, Mao ZW. Platinum-Metformin Conjugates Acting as Promising PD-L1 Inhibitors through the AMP-Activated Protein Kinase Mediated Lysosomal Degradation Pathway. Angew Chem Int Ed Engl 2024:e202410586. [PMID: 39206686 DOI: 10.1002/anie.202410586] [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: 06/05/2024] [Revised: 07/31/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
With the development of metalloimmunology, the potential of platinum drugs in cancer immunotherapy has attracted extensive attention. Although immunochemotherapy combining PD-1/PD-L1 antibodies with platinum drugs has achieved great success in the clinic, combination therapy commonly brings new problems. Herein, we have developed a platinum-metformin conjugate as a promising alternative to antibody-based PD-L1 inhibitors, not only disrupting PD-1/PD-L1 axis on cell surface but also down-regulating the total PD-L1 levels in non-small cell lung cancer (NSCLC) cells comprehensively, thus achieving highly efficient immunochemotherapy by a single small molecule. Mechanism studies demonstrate that Pt-metformin conjugate can selectively accumulate in lysosomes, promote lysosomal-dependent PD-L1 degradation via the AMPK-TFEB pathway, and modulate the upstream regulatory proteins related to PD-L1 expression (e.g. HIF-1α and NF-κB), eventually decreasing the total abundance of PD-L1 in NSCLC, overcoming tumor hypoxia, and activating anti-tumor immunity in vivo. This work suggests an AMPK-mediated lysosomal degradation pathway of PD-L1 for the first time and provides a unique design perspective for the development of novel platinum drugs for immunochemotherapy.
Collapse
Affiliation(s)
- Bin Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Bing-Bing Liang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Wan-Di Cao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Xu-Xian Su
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Qian Cao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| |
Collapse
|
4
|
Man X, Li S, Xu G, Li W, Zhu M, Zhang Z, Liang H, Yang F. Developing a Copper(II) Isopropyl 2-Pyridyl Ketone Thiosemicarbazone Compound Based on the IB Subdomain of Human Serum Albumin-Indomethacin Complex: Inhibiting Tumor Growth by Remodeling the Tumor Microenvironment. J Med Chem 2024; 67:5744-5757. [PMID: 38553427 DOI: 10.1021/acs.jmedchem.3c02378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
To develop a next-generation metal agent and dual-agent multitargeted combination therapy, we developed a copper (Cu) compound based on the properties of the human serum albumin (HSA)-indomethacin (IND) complex to remodel the tumor microenvironment (TME). We optimized a series of Cu(II) isopropyl 2-pyridyl ketone thiosemicarbazone compounds to obtain a Cu(II) compound (C4) with significant cytotoxicity and then constructed an HSA-IND-C4 complex (HSA-IND-C4) delivery system. IND and C4 bind to the hydrophobic cavities of the IB and IIA domains of HSA, respectively. In vivo, the HSA-IND-C4 not only showed enhanced antitumor efficacy relative to C4 and C4 + IND but also improved their targeting ability and decreased their side effects. The antitumor mechanism of C4 + IND involved acting on the different components of the TME. IND inhibited tumor-related inflammation, while C4 not only induced apoptosis and autophagy of cancer cells but also inhibited tumor angiogenesis.
Collapse
Affiliation(s)
- Xueyu Man
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| |
Collapse
|
5
|
Caligiuri R, Massai L, Geri A, Ricciardi L, Godbert N, Facchetti G, Lupo MG, Rossi I, Coffetti G, Moraschi M, Sicilia E, Vigna V, Messori L, Ferri N, Mazzone G, Aiello I, Rimoldi I. Cytotoxic Pt(II) complexes containing alizarin: a selective carrier for DNA metalation. Dalton Trans 2024; 53:2602-2618. [PMID: 38223973 DOI: 10.1039/d3dt03889k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Many efforts have been made in the last few decades to selectively transport antitumor agents to their potential target sites with the aim to improve efficacy and selectivity. Indeed, this aspect could greatly improve the beneficial effects of a specific anticancer agent especially in the case of orphan tumors like the triple negative breast cancer. A possible strategy relies on utilizing a protective leaving group like alizarin as the Pt(II) ligand to reduce the deactivation processes of the pharmacophore enacted by Pt resistant cancer cells. In this study a new series of neutral mixed-ligand Pt(II) complexes bearing alizarin and a variety of diamine ligands were synthesized and spectroscopically characterized by FT-IR, NMR and UV-Vis analyses. Three Pt(II) compounds, i.e., 2b, 6b and 7b, emerging as different both in terms of structural properties and cytotoxic effects (not effective, 10.49 ± 1.21 μM and 24.5 ± 1.5 μM, respectively), were chosen for a deeper investigation of the ability of alizarin to work as a selective carrier. The study comprises the in vitro cytotoxicity evaluation against triple negative breast cancer cell lines and ESI-MS interaction studies relative to the reaction of the selected Pt(II) complexes with model proteins and DNA fragments, mimicking potential biological targets. The results allow us to suggest the use of complex 6b as a prospective anticancer agent worthy of further investigations.
Collapse
Affiliation(s)
- Rossella Caligiuri
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Ponte Pietro Bucci Cubo 14C, Arcavacata di Rende (CS), 87036, Italy.
| | - Lara Massai
- Department of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Andrea Geri
- Department of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Loredana Ricciardi
- CNR-Nanotec, UoS di Cosenza, Dipartimento di Fisica, Università della Calabria, 87036 Rende (CS), Italy
| | - Nicolas Godbert
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Ponte Pietro Bucci Cubo 14C, Arcavacata di Rende (CS), 87036, Italy.
- LPM-Laboratorio Preparazione Materiali, STAR-Lab, Università della Calabria, Via Tito Flavio, 87036 Rende (CS), Italy
| | - Giorgio Facchetti
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milano, Italy.
| | | | - Ilaria Rossi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Italy
| | - Giulia Coffetti
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milano, Italy.
| | - Martina Moraschi
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milano, Italy.
| | - Emilia Sicilia
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Ponte Pietro Bucci Cubo 14C, Arcavacata di Rende (CS), 87036, Italy.
| | - Vincenzo Vigna
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Ponte Pietro Bucci Cubo 14C, Arcavacata di Rende (CS), 87036, Italy.
| | - Luigi Messori
- Department of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Nicola Ferri
- Department of Medicine, University of Padova, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Gloria Mazzone
- Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Ponte Pietro Bucci Cubo 14C, Arcavacata di Rende (CS), 87036, Italy.
| | - Iolinda Aiello
- MAT-INLAB, LASCAMM CR-INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Ponte Pietro Bucci Cubo 14C, Arcavacata di Rende (CS), 87036, Italy.
- LPM-Laboratorio Preparazione Materiali, STAR-Lab, Università della Calabria, Via Tito Flavio, 87036 Rende (CS), Italy
- CNR-Nanotec, UoS di Cosenza, Dipartimento di Fisica, Università della Calabria, 87036 Rende (CS), Italy
| | - Isabella Rimoldi
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milano, Italy.
| |
Collapse
|
6
|
Kastner A, Mendrina T, Babu T, Karmakar S, Poetsch I, Berger W, Keppler BK, Gibson D, Heffeter P, Kowol CR. Stepwise optimization of tumor-targeted dual-action platinum(iv)-gemcitabine prodrugs. Inorg Chem Front 2024; 11:534-548. [PMID: 38235273 PMCID: PMC10790623 DOI: 10.1039/d3qi02032k] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/28/2023] [Indexed: 01/19/2024]
Abstract
While platinum-based chemotherapeutic agents have established themselves as indispensable components of anticancer therapy, they are accompanied by a variety of side effects and the rapid occurrence of drug resistance. A promising strategy to address these challenges is the use of platinum(iv) prodrugs, which remain inert until they reach the tumor tissue, thereby mitigating detrimental effects on healthy cells. Typically, platinum drugs are part of combination therapy settings. Consequently, a very elegant strategy is the development of platinum(iv) prodrugs bearing a second, clinically relevant therapeutic in axial position. In the present study, we focused on gemcitabine as an approved antimetabolite, which is highly synergistic with platinum drugs. In addition, to increase plasma half-life and facilitate tumor-specific accumulation, an albumin-binding maleimide moiety was attached. Our investigations revealed that maleimide-cisplatin(iv)-gemcitabine complexes cannot carry sufficient amounts of gemcitabine to induce a significant effect in vivo. Consequently, we designed a carboplatin(iv) analog, that can be applied at much higher doses. Remarkably, this novel analog demonstrated impressive in vivo results, characterized by significant improvements in overall survival. Notably, these encouraging results could also be transferred to an in vivo xenograft model with acquired gemcitabine resistance, indicating the high potential of this approach.
Collapse
Affiliation(s)
- Alexander Kastner
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- University of Vienna, Vienna Doctoral School in Chemistry (DoSChem) Waehringer Str. 42 1090 Vienna Austria
| | - Theresa Mendrina
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
| | - Tomer Babu
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Subhendu Karmakar
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Isabella Poetsch
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
| | - Walter Berger
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Bernhard K Keppler
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Petra Heffeter
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Christian R Kowol
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| |
Collapse
|
7
|
Liu Z, Cai J, Jiang G, Wang M, Wu C, Su K, Hu W, Huang Y, Yu C, Huang X, Cao G, Wang H. Novel Platinum(IV) complexes intervene oxaliplatin resistance in colon cancer via inducing ferroptosis and apoptosis. Eur J Med Chem 2024; 263:115968. [PMID: 37995563 DOI: 10.1016/j.ejmech.2023.115968] [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: 09/19/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Platinum-based chemotherapeutics are widely used for cancer treatment but are frequently limited because of dosage-dependent side effects and drug resistance. To attenuate these drawbacks, a series of novel platinum(IV) prodrugs (15a-18c) were synthesized and evaluated for anti-cancer activity. Among them, 17a demonstrated superior anti-proliferative activity compared with oxaliplatin (OXA) in the cisplatin-resistant lung cancer cell line A549/CDDP and OXA-resistant colon cancer cell line HCT-116/OXA but showed a lower cytotoxic effect toward human normal cell lines HUVEC and L02. Mechanistic investigations suggested that 17a efficiently enhanced intracellular platinum accumulation, induced DNA damage, disturbed the homeostasis of intracellular reactive oxygen molecules and mitochondrial membrane potential, and thereby activated the mitochondrion-dependent apoptosis pathway. Moreover, 17a significantly induced ferroptosis in HCT-116/OXA via triggering the accumulation of lipid peroxides, disrupting iron homeostasis, and inhibiting solute carrier family 7 member 11 and glutathione peroxidase 4 axial pathway transduction by inhibiting the expression of the phosphorylated signal transducer and activator of transcription 3 and nuclear factor erythroid 2-related factor 2. Moreover, 17a exerted remarkable in vivo antitumor efficacy in the HCT-116/OXA xenograft models but showed attenuated toxicity. These results indicated that these novel platinum(IV) complexes provided an alternative strategy to develop novel platinum-based antineoplastic agents for cancer treatment.
Collapse
Affiliation(s)
- Zhikun Liu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Jinyuan Cai
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Guiyang Jiang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Meng Wang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China
| | - Chuang Wu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Kangning Su
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Weiwei Hu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Yaxian Huang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Chunhao Yu
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Xiaochao Huang
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China.
| | - Guoxiu Cao
- Green Chemistry and Process Enhancement Technology, National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huai'an, 223003, China.
| | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin, 541004, China.
| |
Collapse
|
8
|
Wang R, Li N, Zhang T, Sun Y, He X, Lu X, Chu L, Sun K. Tumor microenvironment-responsive micelles assembled from a prodrug of mitoxantrone and 1-methyl tryptophan for enhanced chemo-immunotherapy. Drug Deliv 2023; 30:2182254. [PMID: 36840464 PMCID: PMC9970211 DOI: 10.1080/10717544.2023.2182254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
Mitoxantrone (MX) can induce the immunogenic-cell death (ICD) of tumor cells and activate anti-tumor immune responses. However, it can also cause high expression of indole amine 2, 3-dioxygenase (IDO) during ICD, leading to T-cell apoptosis and a weakened immune response. An IDO inhibitor, 1-methyl tryptophan (1-MT), can inhibit the activity of IDO caused by MX, resulting in enhanced chemo-immunotherapy. Here, MX-1-MT was connected by ester bond which could be broken in an acidic tumor microenvironment. MX-1-MT was combined with polyethylene glycol (PEG) via a disulfide bond which could be reduced by glutathione overexpressed in tumors, thereby accelerating drug release at target sites. Folic acid-modified distearoyl phosphoethanolamine-polyethylene glycol (DSPE-PEG-FA) was introduced to form targeting micelles. The micelles were of uniform particle size, high stability, and high responsiveness. They could be taken-up by drug-resistant MCF-7/ADR cells, displayed high targeting ability, and induced enhanced cytotoxicity and ICD. Due to 1-MT addition, micelles could inhibit IDO. In vivo studies demonstrated that micelles could accumulate in the tumor tissues of nude mice, resulting in an enhanced antitumor effect and few side-effects.
Collapse
Affiliation(s)
- Ru Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Nuannuan Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Tianyu Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Yiying Sun
- Yantai Saipute Analyzing Service Co. Ltd, Yantai, Shandong Province, China
| | - Xiaoyan He
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Xiaoyan Lu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Liuxiang Chu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Kaoxiang Sun
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China,CONTACT Kaoxiang Sun Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| |
Collapse
|
9
|
Zhu M, Man X, Tongfu Y, Li W, Li S, Xu G, Zhang Z, Liang H, Yang F. Developing a Hetero-Trinuclear Erbium(III)-Copper(II) Complex Based on Apoferritin: Targeted Photoacoustic Imaging and Multimodality Therapy of Tumor. J Med Chem 2023; 66:15424-15436. [PMID: 37956097 DOI: 10.1021/acs.jmedchem.3c01583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
For the integration of targeted diagnosis and treatment of tumor, we innovatively designed and synthesized a single-molecule hetero-multinuclear Er(III)-Cu(II) complex (ErCu2) and then constructed an ErCu2@apoferritin (AFt) nanoparticle (NP) delivery system. ErCu2 and ErCu2@AFt NPs not only provided an evident photoacoustic imaging (PAI) signal of the tumor but also effectively inhibited tumor growth by integrating photothermal therapy, chemotherapy, and immunotherapy. ErCu2@AFt NPs improved the targeting ability and decreased the systemic toxicity of ErCu2 in vivo. Furthermore, we confirmed that ErCu2 and ErCu2@AFt NPs inhibited tumor growth by inducing apoptosis and autophagy of tumor cells and activating the immune system. The study not only provides a novel strategy to develop therapeutic metal agents but also reveals their potential for targeted accurate diagnosis and multimodality therapy of cancer.
Collapse
Affiliation(s)
- Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Xueyu Man
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563006, China
| | - Yang Tongfu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| |
Collapse
|
10
|
Kastner A, Schueffl H, Yassemipour PA, Keppler BK, Heffeter P, Kowol CR. Einbau von (Bioaktiven) Äquatorialen Liganden in Platin(IV)-Komplexe. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 135:e202311468. [PMID: 38516539 PMCID: PMC10952677 DOI: 10.1002/ange.202311468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Indexed: 03/23/2024]
Abstract
AbstractPlatin(IV)‐Prodrugs sind aufgrund ihrer erhöhten Tumorselektivität und geringeren Nebenwirkungen äußerst interessante Alternativen zu Platin(II)‐Antitumortherapeutika. Im Gegensatz zur gängigen Theorie haben wir kürzlich beobachtet, dass äquatoriale Liganden von z. B. Oxaliplatin(IV)‐Komplexen unter Bildung von [(DACH)Pt(OHeq)2(OAcax)2] hydrolysiert werden können. In der hier vorgestellten Arbeit untersuchten wir die Reaktivität und synthetische Verwendbarkeit dieses Komplexes, als Vorstufe für die Entwicklung neuartiger Platin(IV)‐Komplexe, welche mit herkömmlichen Methoden nicht zugänglich sind. Tatsächlich war es möglich die äquatorialen Hydroxidoliganden z. B. durch ein oder zwei monodentate Biotin‐Liganden, die unter Standardmethoden oxidiert werden würden, zu ersetzen. Die gebildeten Komplexe erwiesen sich als sehr stabil und zeigten auch nach der Reduktion eine langsame Ligandenfreisetzung, eine ideale Eigenschaft für lang zirkulierende zielgerichtete Strategien. Daraufhin wurden zwei Platin(IV)‐Komplexe mit äquatorialen Maleimiden, für die Bindung an Serumalbumin als natürlichen Nanocarrier, synthetisiert. Die Komplexe zeigten im Vergleich zu Oxaliplatin eine stark verlängerte Plasmahalbwertszeit und eine deutlich verbesserte Antitumoraktivität in vivo. Zusammenfassend ermöglicht diese neu entwickelte Syntheseplattform den einfachen und gezielten Einbau äquatorialer Liganden in Platin(IV)‐Komplexe. Des Weiteren können verschiedene (bioaktive) Einheiten koordiniert werden, wodurch sogar zielgerichtete dreifach‐wirksame Platin(IV)‐Prodrugs mit nur einem Platinzentrum möglich wären.
Collapse
Affiliation(s)
- Alexander Kastner
- Universität WienFakultät für ChemieInstitut für Anorganische ChemieWähringer Str. 421090WienÖsterreich
- Universität WienVienna Doctoral School in Chemistry (DoSChem)Währinger Str. 421090WienÖsterreich
| | - Hemma Schueffl
- Zentrum für Krebsforschung und Comprehensive Cancer CenterMedizinische Universität WienBorschkegasse 8a1090WienÖsterreich
| | - Patrick A. Yassemipour
- Universität WienFakultät für ChemieInstitut für Anorganische ChemieWähringer Str. 421090WienÖsterreich
| | - Bernhard K. Keppler
- Universität WienFakultät für ChemieInstitut für Anorganische ChemieWähringer Str. 421090WienÖsterreich
- Research Cluster “Translational Cancer Therapy Research”1090WienÖsterreich
| | - Petra Heffeter
- Zentrum für Krebsforschung und Comprehensive Cancer CenterMedizinische Universität WienBorschkegasse 8a1090WienÖsterreich
- Research Cluster “Translational Cancer Therapy Research”1090WienÖsterreich
| | - Christian R. Kowol
- Universität WienFakultät für ChemieInstitut für Anorganische ChemieWähringer Str. 421090WienÖsterreich
- Research Cluster “Translational Cancer Therapy Research”1090WienÖsterreich
| |
Collapse
|
11
|
Kastner A, Schueffl H, Yassemipour PA, Keppler BK, Heffeter P, Kowol CR. Insertion of (Bioactive) Equatorial Ligands into Platinum(IV) Complexes. Angew Chem Int Ed Engl 2023; 62:e202311468. [PMID: 37703130 PMCID: PMC10952260 DOI: 10.1002/anie.202311468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 09/15/2023]
Abstract
Platinum(IV) prodrugs are highly interesting alternatives to platinum(II) anticancer therapeutics due to their increased tumor selectivity and reduced side effects. In contrast to the established theory, we recently observed that the equatorial ligand(s) of e.g. oxaliplatin(IV) complexes can be hydrolyzed with formation of [(DACH)Pt(OHeq )2 (OAcax )2 ]. In the work presented here, we investigated the reactivity and synthetic usability of this complex to be exploited as a precursor for the development of novel platinum(IV) complexes, not able to be synthesized by conventional protocols. Indeed, we could substitute the equatorial hydroxido ligand(s) e.g. by one or two monodentate biotin ligands (which would be oxidized under standard methods). The formed complexes turned out to be very stable with slow ligand release after reduction, ideal for long-circulating tumor-targeting strategies. Therefore, two platinum(IV) complexes with equatorial maleimides, capable of exploiting serum albumin as a natural nanocarrier, were synthesized as well. The complexes showed massively prolonged plasma half-life and distinctly improved anticancer activity in vivo compared to oxaliplatin. Taken together, the newly developed synthetic platform allows the simple and specific insertion of equatorial ligands into platinum(IV) complexes. This will enable the attachment of three different (bioactive) moieties generating targeted triple-action platinum(IV) prodrugs within one single platinum complex.
Collapse
Affiliation(s)
- Alexander Kastner
- University of ViennaFaculty of ChemistryInstitute of Inorganic ChemistryWaehringer Str. 421090ViennaAustria
- University of ViennaVienna Doctoral School in Chemistry (DoSChem)Waehringer Str. 421090ViennaAustria
| | - Hemma Schueffl
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaBorschkegasse 8a1090ViennaAustria
| | - Patrick A. Yassemipour
- University of ViennaFaculty of ChemistryInstitute of Inorganic ChemistryWaehringer Str. 421090ViennaAustria
| | - Bernhard K. Keppler
- University of ViennaFaculty of ChemistryInstitute of Inorganic ChemistryWaehringer Str. 421090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”1090ViennaAustria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of ViennaBorschkegasse 8a1090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”1090ViennaAustria
| | - Christian R. Kowol
- University of ViennaFaculty of ChemistryInstitute of Inorganic ChemistryWaehringer Str. 421090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”1090ViennaAustria
| |
Collapse
|
12
|
Wang M, Cao G, Zhou J, Cai J, Ma X, Liu Z, Huang X, Wang H. Ligustrazine-Derived Chalcones-Modified Platinum(IV) Complexes Intervene in Cisplatin Resistance in Pancreatic Cancer through Ferroptosis and Apoptosis. J Med Chem 2023; 66:13587-13606. [PMID: 37766483 DOI: 10.1021/acs.jmedchem.3c00922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Developing multitarget platinum(IV) prodrugs is an important strategy to attenuate cisplatin (CDDP) resistance in tandem with reduced toxicity. Herein, six novel ligustrazine-derived chalcones-modified platinum(IV) complexes were synthesized and evaluated for their anti-proliferative activities. Among them, 16a displayed higher cytotoxicity toward the tested cancer cell lines and lower cytotoxicity toward the human normal cells than CDDP or the combined group. Mechanistic studies revealed that 16a efficiently induced DNA damage and initiated a mitochondria-dependent apoptosis pathway. Besides, 16a significantly triggered ferroptosis by down-regulating expression levels of nuclear factor erythroid 2-related factor 2, glutathione peroxidase 4, and solute carrier family 7 member 11. Further, 16a obtained superior in vivo anti-tumor efficiency than CDDP in CDDP-resistant pancreatic cancer xenograft models but showed no significant side effects. In summary, our study suggested that 16a acts via a different anti-cancer mechanistic pathway than CDDP and may therefore encompass a novel practical strategy for cancer treatment.
Collapse
Affiliation(s)
- Meng Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Guoxiu Cao
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Junjie Zhou
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Jinyuan Cai
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Xianjie Ma
- Department of Pharmacy, People's Hospital of Rizhao, Rizhao 276827, China
| | - Zhikun Liu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Xiaochao Huang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
13
|
Chen Y, Li L, Liu Z, Liu M, Wang Q. A series of ligustrazine platinum(IV) complexes with potent anti-proliferative and anti-metastatic properties that exert chemotherapeutic and immunotherapeutic effects. Dalton Trans 2023; 52:13097-13109. [PMID: 37664893 DOI: 10.1039/d3dt02358c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The development of novel anticancer drugs with antiproliferative and antimetastatic activities is of great importance in the pharmaceutical field. Herein, a series of ligustrazine (LSZ) platinum(IV) complexes with chemotherapeutic and immunotherapeutic effects were designed, prepared and evaluated as antitumor agents for the first time. Complex 4 with potent antitumor activities both in vitro and in vivo was screened out as a candidate. Notably, it displays significantly more effective anti-metastatic activities than the platinum(II) drugs cisplatin and oxaliplatin. Mechanism detection discloses that it causes serious DNA damage and increases the expression of γ-H2AX and P53. Then, the apoptosis of tumor cells is promoted by activating the mitochondrial apoptotic pathway Bcl-2/Bax/caspase-3 and causing autophagy via modulating LC3-I/II and P62 expression. Furthermore, the immune therapeutic responses are significantly elevated by blocking HIF-1α, ERK 1/2 and COX-2 pathways to reduce PD-L1 expression, and further increasing CD3+ and CD8+ T cells to elevate T cell immunity in tumors. Tumor metastasis is blocked by the synergistic functions of DNA damage, hypoxia modulation and immune activation.
Collapse
Affiliation(s)
- Yan Chen
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Linming Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| | - Meifeng Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P.R. China.
| |
Collapse
|
14
|
Vinck R, Dömötör O, Karges J, Jakubaszek M, Seguin J, Tharaud M, Guérineau V, Cariou K, Mignet N, Enyedy ÉA, Gasser G. In Situ Bioconjugation of a Maleimide-Functionalized Ruthenium-Based Photosensitizer to Albumin for Photodynamic Therapy. Inorg Chem 2023; 62:15510-15526. [PMID: 37708255 DOI: 10.1021/acs.inorgchem.3c01984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Maleimide-containing prodrugs can quickly and selectively react with circulating serum albumin following their injection in the bloodstream. The drug-albumin complex then benefits from longer blood circulation times and better tumor accumulation. Herein, we have applied this strategy to a previously reported highly phototoxic Ru polypyridyl complex-based photosensitizer to increase its accumulation at the tumor, reduce off-target cytotoxicity, and therefore improve its pharmacological profile. Specifically, two complexes were synthesized bearing a maleimide group: one complex with the maleimide directly incorporated into the bipyridyl ligand, and the other has a hydrophilic linker between the ligand and the maleimide group. Their interaction with albumin was studied in-depth, revealing their ability to efficiently bind both covalently and noncovalently to the plasma protein. A crucial finding is that the maleimide-functionalized complexes exhibited significantly lower cytotoxicity in noncancerous cells under dark conditions compared to the nonfunctionalized complex, which is a highly desirable property for a photosensitizer. The binding to albumin also led to a decrease in the phototoxicity of the Ru bioconjugates in comparison to the nonfunctionalized complex, probably due to a decreased cellular uptake. Unfortunately, this decrease in phototoxicity was not compensated by a dramatic increase in tumor accumulation, as was demonstrated in a tumor-bearing mouse model using inductively coupled plasma mass spectrometry (ICP-MS) studies. Consequently, this study provides valuable insight into the future design of in situ albumin-binding complexes for photodynamic therapy in order to maximize their effectiveness and realize their full potential.
Collapse
Affiliation(s)
- Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Orsolya Dömötör
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7. H-6720 Szeged, Hungary
| | - Johannes Karges
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Marta Jakubaszek
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Johanne Seguin
- Université Paris Cité, UTCBS, INSERM, CNRS, 75006 Paris, France
| | - Mickaël Tharaud
- Biogéochimie à l'Anthropocène des Eléments et Contaminants Emergents, Institut de Physique du Globe de Paris, 75005 Paris, France
| | - Vincent Guérineau
- Institut de Chimie des Substances Naturelles, CNRS UPR2301, Université Paris-Sud, Université Paris-Saclay, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cedex, France
| | - Kevin Cariou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| | - Nathalie Mignet
- Université Paris Cité, UTCBS, INSERM, CNRS, 75006 Paris, France
| | - Éva A Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7. H-6720 Szeged, Hungary
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, F-75005 Paris, France
| |
Collapse
|
15
|
Hao Y, Li R, Pan W, Tian S, Min Y. Platinum Twin and Triplet Drugs Improve Chemoimmunotherapy. J Med Chem 2023; 66:12225-12236. [PMID: 37665669 DOI: 10.1021/acs.jmedchem.3c00792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Several chemoimmunotherapy regimens have been approved by the U.S. FDA, verifying the great clinical value and potential of the strategy. However, the immunomodulatory function of chemotherapy was insufficient, which did not provide extra overall survival benefits, especially in a head-to-head comparison of chemoimmunotherapy versus immunotherapy. Here, we engineered twin and triplet drugs derived from an immunogenic chemotherapeutic drug (oxaliplatin) and small-molecule inhibitors of negative immunoregulation pathways (COX2 and IDO) in tumors as an improved chemotherapeutic component within chemoimmunotherapy. The twin and triplet drugs exhibited significantly improved synergy with anti-PD-1 in a CT26 colorectal mouse tumor model. Mechanistic analyses revealed that the drug induced immunogenic cell death and restored tumor immune microenvironment toward tumor clearance in vivo, resulting in a great decrease in tumor-infiltrating Tregs and an increase in the CD8+ T/Treg ratio when combined with anti-PD-1. Our work expands the application of platinum twin drugs in combination with an immune checkpoint blockade.
Collapse
Affiliation(s)
- Yuhao Hao
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Rui Li
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Wen Pan
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Shaomin Tian
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290, United States
| | - Yuanzeng Min
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei 230031, China
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
16
|
Kastner A, Mendrina T, Bachmann F, Berger W, Keppler BK, Heffeter P, Kowol CR. Tumor-targeted dual-action NSAID-platinum(iv) anticancer prodrugs. Inorg Chem Front 2023; 10:4126-4138. [PMID: 37440920 PMCID: PMC10334471 DOI: 10.1039/d3qi00968h] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023]
Abstract
Platinum(iv) prodrugs are a promising class of anticancer agents designed to overcome the limitations of conventional platinum(ii) therapeutics. In this work, we present oxaliplatin(iv)-based complexes, which upon reduction, release acetylsalicylic acid (aspirin), known for its antitumor activity against colon cancer and currently investigated in combination with oxaliplatin in a phase III clinical study. Comparison with a recently reported cisplatin analog (asplatin) revealed a massive increase in reduction stability for the oxaliplatin complex in mouse serum. This was in line with the cell culture data indicating the desired prodrug properties for the newly synthesized complex. For in vivo studies, a new derivative containing an albumin-binding maleimide unit was synthesized. Indeed, distinctly longer plasma half-life as well as higher tumor accumulation in comparison to asplatin and oxaliplatin were observed, also leading to significantly higher antitumor activity and overall survival of CT26 tumor-bearing mice.
Collapse
Affiliation(s)
- Alexander Kastner
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- University of Vienna, Vienna Doctoral School in Chemistry (DoSChem) Waehringer Str. 42 1090 Vienna Austria
| | - Theresa Mendrina
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Florian Bachmann
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
| | - Walter Berger
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Bernhard K Keppler
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Petra Heffeter
- Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna Borschkegasse 8a 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| | - Christian R Kowol
- University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry Waehringer Str. 42 1090 Vienna Austria
- Research Cluster "Translational Cancer Therapy Research" 1090 Vienna Austria
| |
Collapse
|
17
|
Coffetti G, Moraschi M, Facchetti G, Rimoldi I. The Challenging Treatment of Cisplatin-Resistant Tumors: State of the Art and Future Perspectives. Molecules 2023; 28:molecules28083407. [PMID: 37110640 PMCID: PMC10144581 DOI: 10.3390/molecules28083407] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
One of the main problems in chemotherapy using platinum drugs as anticancer agents is the resistance phenomenon. Synthesizing and evaluating valid alternative compounds is challenging. This review focuses on the last two years of progress in the studies of platinum (II)- and platinum (IV)-based anticancer complexes. In particular, the research studies reported herein focus on the capability of some platinum-based anticancer agents to bypass resistance to chemotherapy, which is typical of well-known drugs such as cisplatin. Regarding platinum (II) complexes, this review deals with complexes in trans conformation; complexes containing bioactive ligands, as well as those that are differently charged, all experience a different reaction mechanism compared with cisplatin. Regarding platinum (IV) compounds, the focus was on complexes with biologically active ancillary ligands that exert a synergistic effect with platinum (II)-active complexes upon reduction, or those for which controllable activation can be realized thanks to intracellular stimuli.
Collapse
Affiliation(s)
- Giulia Coffetti
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milano, Italy
| | - Martina Moraschi
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milano, Italy
| | - Giorgio Facchetti
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milano, Italy
| | - Isabella Rimoldi
- Department of Pharmaceutical Sciences, University of Milan, Via Venezian 21, 20133 Milano, Italy
| |
Collapse
|
18
|
Li R, Zhao W, Jin C, Xiong H. Dual-target platinum(IV) complexes reverse cisplatin resistance in triple negative breast via inhibiting poly(ADP-ribose) polymerase (PARP-1) and enhancing DNA damage. Bioorg Chem 2023; 133:106354. [PMID: 36720184 DOI: 10.1016/j.bioorg.2023.106354] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/29/2022] [Accepted: 01/08/2023] [Indexed: 01/19/2023]
Abstract
Platinum(II)-based drugs play an important role in many chemotherapeutic protocols, but their further clinical applications are hindered by the development of drug resistance and serious side effects. Therefore, to reverse cisplatin (CDDP) resistance in tandem with reduced side effects, nine novel platinum(IV) complexes modified with key pharmacophore of Olaparib were synthesized and evaluated for biological activities. Among them, the optimal complex 8-2 showed good inhibitory activity against PARP-1 and superior anticancer effects over CDDP on parental (MDA-MB-231, IC50 = 1.13 μM) and CDDP -resistant triple-negative breast cancer (TNBC) cell line (MDA-MB-231/CDDP, IC50 = 1.72 μM). Detailed mechanisms revealed that compared with Olaparib and CDDP, the enhanced intracellular accumulation of 8-2 could efficiently reverse CDDP resistance in MDA-MB-231/CDDP cells via inhibiting DNA repair-associated mechanisms, enhancing DNA damage, and activating mitochondrion-dependent apoptosis pathway. Furthermore, 8-2 obtained higher tumor growth inhibition rate (64.1 %) than CDDP (26.5 %) in MDA-MB-231/CDDP xenografts, but it did not induce significant toxicity in vivo and in intro, making it a potential drug candidate for the treatment of TNBC.
Collapse
Affiliation(s)
- Rui Li
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chen Jin
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
19
|
Cao X, Li R, Wang H, Guo C, Wang S, Chen X, Zhao R. Novel indole–chalcone platinum(IV) complexes as tubulin polymerization inhibitors to overcome oxaliplatin resistance in colorectal cancer. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
20
|
Yao H, Zhu G. Blood Components as Carriers for Small-Molecule Platinum Anticancer Drugs. ChemMedChem 2022; 17:e202200482. [PMID: 36178204 DOI: 10.1002/cmdc.202200482] [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: 09/05/2022] [Revised: 09/29/2022] [Indexed: 02/01/2023]
Abstract
The efficacy of platinum drugs is limited by severe side effects, drug resistance, and poor pharmacokinetic properties. Utilizing long-lasting blood components as drug carriers is a promising strategy to improve the circulation half-lives and tumor accumulation of platinum drugs. Non-immunogenic blood cells such as erythrocytes and blood proteins such as albumins, which have long lifespans, are suitable for the delivery of platinum drugs. In this concept, we briefly summarize the strategies of applying blood components as promising carriers to deliver small-molecule platinum drugs for cancer treatment. Examples of platinum drugs that are encapsulated, non-covalently attached, and covalently bound to erythrocytes and plasma proteins such as albumin and apoferritin are introduced. The potential methods to increase the stability of platinum-based thiol-maleimide conjugates involved in these delivery systems are also discussed. This concept may enlighten researchers with more ideas on the future development of novel platinum drugs that have excellent pharmacokinetic properties and antitumor performance in vivo.
Collapse
Affiliation(s)
- Houzong Yao
- School of Health, Jiangxi Normal University, Nanchang, 330022, P. R. China.,Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong, SAR, 999077, P. R. China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong, SAR, 999077, P. R. China.,City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, P. R. China
| |
Collapse
|
21
|
Integrative Metallomics Studies of Toxic Metal(loid) Substances at the Blood Plasma–Red Blood Cell–Organ/Tumor Nexus. INORGANICS 2022. [DOI: 10.3390/inorganics10110200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Globally, an estimated 9 million deaths per year are caused by human exposure to environmental pollutants, including toxic metal(loid) species. Since pollution is underestimated in calculations of the global burden of disease, the actual number of pollution-related deaths per year is likely to be substantially greater. Conversely, anticancer metallodrugs are deliberately administered to cancer patients, but their often dose-limiting severe adverse side-effects necessitate the urgent development of more effective metallodrugs that offer fewer off-target effects. What these seemingly unrelated events have in common is our limited understanding of what happens when each of these toxic metal(loid) substances enter the human bloodstream. However, the bioinorganic chemistry that unfolds at the plasma/red blood cell interface is directly implicated in mediating organ/tumor damage and, therefore, is of immediate toxicological and pharmacological relevance. This perspective will provide a brief synopsis of the bioinorganic chemistry of AsIII, Cd2+, Hg2+, CH3Hg+ and the anticancer metallodrug cisplatin in the bloodstream. Probing these processes at near-physiological conditions and integrating the results with biochemical events within organs and/or tumors has the potential to causally link chronic human exposure to toxic metal(loid) species with disease etiology and to translate more novel anticancer metal complexes to clinical studies, which will significantly improve human health in the 21st century.
Collapse
|
22
|
Velcheva V, Hegetschweiler K, Momekov G, Ivanova S, Ugrinov A, Morgenstern B, Gencheva G. Platinum(IV) Complexes of the 1,3,5-Triamino Analogue of the Biomolecule Cis-Inositol Designed as Innovative Antineoplastic Drug Candidates. Pharmaceutics 2022; 14:2057. [PMID: 36297500 PMCID: PMC9611922 DOI: 10.3390/pharmaceutics14102057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 02/01/2023] Open
Abstract
Metal complexes occupy a special place in the field of treatment and diagnostics. Their main advantages stem from the possibility of fine-tuning their thermodynamic properties and kinetic behavior in the biological milieu by applying different approaches such as properly constructed inner coordination sphere, appropriate choice of ligands, metal oxidation state, redox potential, etc., which are specific to these compounds. Here we discuss the design and synthesis of two octahedral cationic Pt(IV) complexes of the tridentate ligand all-cis-2,4,6-triaminocyclohexane-1,3,5-triol (taci) with composition, fac-[Pt(taci)I3]+, 1 and bis-[Pt(taci)2]4+, 2 as well as the potential for their application as antineoplastic agents. The complexes have been isolated in a solid state as: fac-[Pt(taci)I3]I·3H2O (1A), fac-[Pt(taci)I3]I (1B), fac-[Pt(taci)I3]I·2DMF (1C), bis-[Pt(taci)2](CO3)2·6H2O (2A) by changing the acidity of the reaction systems, the molar ratios of the reagents and the counterions, and by re-crystallization. The ligand taci is coordinated through the NH2-groups, each molecule occupying three coordination places in the inner coordination sphere of Pt(IV). Monitoring of the hydrolysis processes of 1A and 2A at different acidity showed that while 2A remained stable over the study period, the I--ions in 1A were successively substituted, with the main product under physiologically mimetic conditions being fac,cis-[Pt(taci)I(OH)2]+ (h2). The antiproliferative tests involved eight cancer cell models, among which chemosensitive (derived from leukemias and solid tumors) and chemoresistant human Acute myeloid leukemia lines (HL-60/Dox, HL-60/CDDP), as well as the non-malignant kidney' cells HEK-293T showed that the complexes 1A and 2A are characterized by a fundamentally different profile of chemosensitivity and spectrum of cytotoxic activity compared to cisplatin. The new Pt(IV) complexes were shown to be more effective in selectively inhibiting the proliferation of human malignant cells compared to cisplatin. Remarkable activity was recorded for 1A, which showed an effect (IC50 = 8.9 ± 2.4) at more than 16-fold lower concentration than cisplatin (IC50 = 144.4 ± 9.8) against the resistant cell line HL-60/CDDP. In parallel, 1A exhibited virtually the same cytotoxic effect against the parental HL-60 cells (IC50 = 9.0 ± 1.2), where cisplatin displays comparable chemosensitivity (IC50 = 8.3 ± 0.8). The determined resistance indices (RI~1) show unequivocally that the resistant lines are sensitive to both compounds tested; therefore, they are capable of overcoming the mechanisms of cisplatin resistance. The structural features of these compounds and their promising pharmacological properties justify their inclusion in the group of "non-classical metal-based antitumor compounds" and are a prerequisite for the admission of alternative mechanisms of action.
Collapse
Affiliation(s)
- Vyara Velcheva
- Faculty of Chemistry and Pharmacy, Sofia University “St. Kliment Ohridski”, 1 J. Bourchier Blvd., 1164 Sofia, Bulgaria
| | - Kaspar Hegetschweiler
- Fachrichtung Chemie, Universität des Saarlandes, Campus, D-66123 Saarbrücken, Germany
| | - Georgi Momekov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
| | - Stefka Ivanova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Medical University of Pleven, 1 St. Kliment Ohridski Str., 5800 Pleven, Bulgaria
| | - Angel Ugrinov
- Department of Chemistry and Biochemistry, North Dakota State University, 1311 Albrecht Blvd., Fargo, ND 58102, USA
| | - Bernd Morgenstern
- Fachrichtung Chemie, Universität des Saarlandes, Campus, D-66123 Saarbrücken, Germany
| | - Galina Gencheva
- Faculty of Chemistry and Pharmacy, Sofia University “St. Kliment Ohridski”, 1 J. Bourchier Blvd., 1164 Sofia, Bulgaria
| |
Collapse
|
23
|
Khosravifarsani M, Ait-Mohand S, Paquette B, Sanche L, Guérin B. In vivo behavior of [64Cu]NOTA-terpyridine platinum, a novel chemo-radio-theranostic agent for imaging, and therapy of colorectal cancer. Front Med (Lausanne) 2022; 9:975213. [PMID: 36226156 PMCID: PMC9549809 DOI: 10.3389/fmed.2022.975213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
To overcome resistance to chemotherapy for colorectal cancer, we propose to validate in vivo a novel terpyridine-platinum (TP) compound radiolabeled with the radio-theranostic isotope 64Cu. In vivo stability, biodistribution, PET imaging, tumor growth delay, toxicity and dosimetry of [64Cu]NOTA-C3-TP were determined. The current experimental studies show that [64Cu]NOTA-C3-TP is stable in vivo, rapidly eliminated by the kidneys and has a promising tumor uptake ranging from 1.8 ± 0.4 to 3.0 ± 0.2 %ID/g over 48 h. [64Cu]NOTA-C3-TP retarded tumor growth by up to 6 ± 2.0 days and improved survival relative to vehicle and non-radioactive [NatCu]NOTA-C3-TP over 17 days of tumor growth observation. This effect was obtained with only 0.4 nmol i.v. injection of [64Cu]NOTA-C3-TP, which delivers 3.4 ± 0.3 Gy tumoral absorbed dose. No evidence of toxicity, by weight loss or mortality was revealed. These findings confirm the high potential of [64Cu]NOTA-TP as a novel radio-theranostic agent.
Collapse
Affiliation(s)
- Meysam Khosravifarsani
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Samia Ait-Mohand
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Benoit Paquette
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Léon Sanche
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Sherbrooke Molecular Imaging Center (CIMS), Centre de Recherche du CHUS (CRCHUS), Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
24
|
Liu F, Yang C, Li S, Wu X, Xue K, Zhou Y, Liang X, Cheng X, Shi Q, Su W. Design and biological features of platinum (II) complexes with 3-hydroxy-3-(Trifluoromethyl)cyclobutane-1,1-Dicarboxylate as a leaving ligand. Eur J Med Chem 2022; 242:114673. [PMID: 36049275 DOI: 10.1016/j.ejmech.2022.114673] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 11/04/2022]
Abstract
A series of platinum compounds 2a-5a and 2b-5b with fluoro-functional groups are designed and synthesized. Among them, complex 2b is the most effective agent with 3-hydroxy-3-(trifluoromethyl)cyclobutane-1,1-dicarboxylate as a leaving ligand, which showed better cytotoxic activity than compounds containing only CF3 or OH group at 3-position of cyclobutane-1,1-dicarboxylate. The water solubility of 2a is better than that of carboplatin (32 mg/mL vs. 16 mg/mL), and its antitumor activity on A549 is 4.6-fold higher than that of carboplatin. The IC50 value of 2b on A549 cells is 4.73 ± 0.64 μM, which is comparable to that of oxaliplatin and higher than that of carboplatin. Meanwhile, 2a and 2b are less toxic than oxaliplatin and cisplatin toward BEAS-2B cells. Moreover, 2a and 2b induce cell apoptosis in vitro by the Bax-Bcl-2-caspase-3 pathway and ferroptosis through inhibiting GPx-4 and elevating COX2. Results from in vivo experiment show that the inhibition rate of A549 xenograft tumor is cisplatin > 2b > oxaliplatin > 2a > carboplatin.
Collapse
Affiliation(s)
- Fengfan Liu
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Chen Yang
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shaoguang Li
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiaoqi Wu
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Keming Xue
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yibo Zhou
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiaobing Liang
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiang Cheng
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Qiwen Shi
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Weike Su
- National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
25
|
Kasparkova J, Kostrhunova H, Novohradsky V, Ma L, Zhu G, Milaeva ER, Shtill AA, Vinck R, Gasser G, Brabec V, Nazarov AA. Is antitumor Pt(IV) complex containing two axial lonidamine ligands a true dual- or multi-action prodrug? METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6618656. [PMID: 35759404 DOI: 10.1093/mtomcs/mfac048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/09/2022] [Indexed: 11/14/2022]
Abstract
This work studied the mechanism of action of a Pt(IV) complex 2 bearing two axial lonidamine ligands, which are selective inhibitors of aerobic glycolysis. The presence of two lonidamine ligands in 2 compared to the parent Pt(II) complex increased its antiproliferative activity, cellular accumulation, and changed its cell cycle profile and mechanism of cell death. In 3D cell culture, 2 showed exceptional antiproliferative activity with IC50 values as low as 1.6 μM in MCF7 cells. The study on the influence of the lonidamine ligands in the Pt complex on glycolysis showed only low potency of ligands to affect metabolic processes in cancer cells, making the investigated complex, not a dual- or multi-action prodrug. However, the Pt(IV) prodrug effectively delivers the cytotoxic Pt(II) complex into cancer cells.
Collapse
Affiliation(s)
- Jana Kasparkova
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Hana Kostrhunova
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Vojtech Novohradsky
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Lili Ma
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P. R. China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P. R. China
| | - Elena R Milaeva
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russian Federation
| | - Alexender A Shtill
- Blokhin Cancer Center, Russian Academy of Medical Sciences, 115478 Moscow, Russian Federation
| | - Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, 75005 Paris, France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, 75005 Paris, France
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Alexey A Nazarov
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russian Federation
| |
Collapse
|
26
|
Han W, He W, Song Y, Zhao J, Song Z, Shan Y, Hua W, Sun Y. Multifunctional platinum(IV) complex bearing HDAC inhibitor and biotin moiety exhibits prominent cytotoxicity and tumor-targeting ability. Dalton Trans 2022; 51:7343-7351. [PMID: 35466968 DOI: 10.1039/d2dt00090c] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite the wide clinical use of platinum drugs in cancer treatment, their severe side effects and lack of tumor selectivity seriously limit their further clinical application. To address the limitations of the current platinum drugs, herein a multifunctional platinum(IV) compound 1 containing a histone deacetylase (HDAC) inhibitor (4-phenylbutyric acid, 4-PBA) and a tumor-targeting group (biotin) has been designed and prepared. An in vitro cytotoxicity study indicated that compound 1 exhibits comparable or superior cytotoxicity to cisplatin against the tested cancer cell lines, but greatly reduced toxicity in human normal liver LO2 cells, implying the potential tumor-targeting ability of compound 1. Molecular docking results indicate that compound 1 can effectively interact with a biotin-specific receptor (streptavidin) through its biotin moiety, enabling potential tumor-targeting capability. Further studies indicated that compound 1's cytotoxicity stems from inducing DNA damage via the mitochondrial apoptotic pathway and inhibiting HDACs. Consequently, this compound can not only take advantage of the tumor selectively of biotin to improve its tumor-targeting ability but also strengthen its anticancer activity via simultaneously targeting DNA and HDACs.
Collapse
Affiliation(s)
- Weinan Han
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Weiyu He
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Yutong Song
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Jian Zhao
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research and Pharmaceutical Research Center, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P.R. China.,Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huai'an 223003, P.R. China
| | - Zhiheng Song
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Yi Shan
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| | - Wuyang Hua
- School of Food Engineering, Jilin Agricultural Science and Technology University, Jilin 132000, P.R. China
| | - Yanyan Sun
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, P.R. China.
| |
Collapse
|
27
|
Ferraro MG, Piccolo M, Misso G, Santamaria R, Irace C. Bioactivity and Development of Small Non-Platinum Metal-Based Chemotherapeutics. Pharmaceutics 2022; 14:pharmaceutics14050954. [PMID: 35631543 PMCID: PMC9147010 DOI: 10.3390/pharmaceutics14050954] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Countless expectations converge in the multidisciplinary endeavour for the search and development of effective and safe drugs in fighting cancer. Although they still embody a minority of the pharmacological agents currently in clinical use, metal-based complexes have great yet unexplored potential, which probably hides forthcoming anticancer drugs. Following the historical success of cisplatin and congeners, but also taking advantage of conventional chemotherapy limitations that emerged with applications in the clinic, the design and development of non-platinum metal-based chemotherapeutics, either as drugs or prodrugs, represents a rapidly evolving field wherein candidate compounds can be fine-tuned to access interactions with druggable biological targets. Moving in this direction, over the last few decades platinum family metals, e.g., ruthenium and palladium, have been largely proposed. Indeed, transition metals and molecular platforms where they originate are endowed with unique chemical and biological features based on, but not limited to, redox activity and coordination geometries, as well as ligand selection (including their inherent reactivity and bioactivity). Herein, current applications and progress in metal-based chemoth are reviewed. Converging on the recent literature, new attractive chemotherapeutics based on transition metals other than platinum—and their bioactivity and mechanisms of action—are examined and discussed. A special focus is committed to anticancer agents based on ruthenium, palladium, rhodium, and iridium, but also to gold derivatives, for which more experimental data are nowadays available. Next to platinum-based agents, ruthenium-based candidate drugs were the first to reach the stage of clinical evaluation in humans, opening new scenarios for the development of alternative chemotherapeutic options to treat cancer.
Collapse
Affiliation(s)
- Maria Grazia Ferraro
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Marialuisa Piccolo
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Gabriella Misso
- Department of Precision Medicine, School of Medicine and Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: (G.M.); (C.I.)
| | - Rita Santamaria
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Carlo Irace
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
- Correspondence: (G.M.); (C.I.)
| |
Collapse
|
28
|
Fronik P, Gutmann M, Vician P, Stojanovic M, Kastner A, Heffeter P, Pirker C, Keppler BK, Berger W, Kowol CR. A platinum(IV) prodrug strategy to overcome glutathione-based oxaliplatin resistance. Commun Chem 2022; 5:46. [PMID: 36697790 PMCID: PMC9814792 DOI: 10.1038/s42004-022-00661-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/07/2022] [Indexed: 02/01/2023] Open
Abstract
Clinical efficacy of oxaliplatin is frequently limited by severe adverse effects and therapy resistance. Acquired insensitivity to oxaliplatin is, at least in part, associated with elevated levels of glutathione (GSH). In this study we report on an oxaliplatin-based platinum(IV) prodrug, which releases L-buthionine-S,R-sulfoximine (BSO), an inhibitor of glutamate-cysteine ligase, the rate-limiting enzyme in GSH biosynthesis. Two complexes bearing either acetate (BSO-OxOAc) or an albumin-binding maleimide (BSO-OxMal) as second axial ligand were synthesized and characterized. The in vitro anticancer activity of BSO-OxOAc was massively reduced in comparison to oxaliplatin, proving its prodrug nature. Nevertheless, the markedly lower intracellular oxaliplatin uptake in resistant HCT116/OxR cells was widely overcome by BSO-OxOAc resulting in distinctly reduced resistance levels. Platinum accumulation in organs of a colorectal cancer mouse model revealed higher tumor selectivity of BSO-OxMal as compared to oxaliplatin. This corresponded with increased antitumor activity, resulting in significantly enhanced overall survival. BSO-OxMal-treated tumors exhibited reduced GSH levels, proliferative activity and enhanced DNA damage (pH2AX) compared to oxaliplatin. Conversely, pH2AX staining especially in kidney cells was distinctly increased by oxaliplatin but not by BSO-OxMal. Taken together, our data provide compelling evidence for enhanced tumor specificity of the oxaliplatin(IV)/BSO prodrug.
Collapse
Affiliation(s)
- Philipp Fronik
- grid.10420.370000 0001 2286 1424University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Michael Gutmann
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Petra Vician
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Mirjana Stojanovic
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Alexander Kastner
- grid.10420.370000 0001 2286 1424University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Petra Heffeter
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Christine Pirker
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Bernhard K. Keppler
- grid.10420.370000 0001 2286 1424University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Waehringer Strasse 42, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Walter Berger
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Christian R. Kowol
- grid.10420.370000 0001 2286 1424University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Waehringer Strasse 42, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| |
Collapse
|
29
|
Barth MC, Lange S, Häfner N, Ueberschaar N, Görls H, Runnebaum IB, Weigand W. Synthesis and characterization of thiocarbonato-linked platinum(IV) complexes. Dalton Trans 2022; 51:5567-5576. [PMID: 35311885 DOI: 10.1039/d2dt00318j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein we show the formation of new oxaliplatin-based platinum(IV) complexes by reaction with DSC-activated thiols via thiocarbonate linkage. Three model complexes based on aliphatic and aromatic thiols, as well as one complex with N-acetylcysteine as biologically active thiol were synthesized. This synthetic strategy affords the expansion of biologically active compounds other than those containing carboxylic, amine or hydroxy groups for coupling to the platinum(IV) center. The complexes were characterized by high-resolution mass spectrometry, NMR spectroscopy (1H, 13C, 195Pt) and elemental analysis. Their biological behavior was evaluated against two ovarian carcinoma cell lines and their cisplatin-resistant analogues. Remarkably, the platinum(IV) samples show modest in vitro cytotoxicity against A2780 cells and comparable effects against A2780cis cells. Two complexes in particular demonstrate improved activity against SKOV3cis cells. The reduction experiment of complex 8, investigated by UHPLC-HRMS, provides evidence of interesting platinum-species formed during reaction with ascorbic acid.
Collapse
Affiliation(s)
- Marie-Christin Barth
- Department of Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 8, 07743 Jena, Germany.
| | - Stefanie Lange
- Department of Gynecology and Reproduction Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Norman Häfner
- Department of Gynecology and Reproduction Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Nico Ueberschaar
- Mass Spectrometry Platform, Friedrich Schiller University Jena, Humboldtstrasse 8, 07743 Jena, Germany
| | - Helmar Görls
- Department of Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 8, 07743 Jena, Germany.
| | - Ingo B Runnebaum
- Department of Gynecology and Reproduction Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Wolfgang Weigand
- Department of Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 8, 07743 Jena, Germany.
| |
Collapse
|
30
|
Peña Q, Wang A, Zaremba O, Shi Y, Scheeren HW, Metselaar JM, Kiessling F, Pallares RM, Wuttke S, Lammers T. Metallodrugs in cancer nanomedicine. Chem Soc Rev 2022; 51:2544-2582. [PMID: 35262108 DOI: 10.1039/d1cs00468a] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metal complexes are extensively used for cancer therapy. The multiple variables available for tuning (metal, ligand, and metal-ligand interaction) offer unique opportunities for drug design, and have led to a vast portfolio of metallodrugs that can display a higher diversity of functions and mechanisms of action with respect to pure organic structures. Clinically approved metallodrugs, such as cisplatin, carboplatin and oxaliplatin, are used to treat many types of cancer and play prominent roles in combination regimens, including with immunotherapy. However, metallodrugs generally suffer from poor pharmacokinetics, low levels of target site accumulation, metal-mediated off-target reactivity and development of drug resistance, which can all limit their efficacy and clinical translation. Nanomedicine has arisen as a powerful tool to help overcome these shortcomings. Several nanoformulations have already significantly improved the efficacy and reduced the toxicity of (chemo-)therapeutic drugs, including some promising metallodrug-containing nanomedicines currently in clinical trials. In this critical review, we analyse the opportunities and clinical challenges of metallodrugs, and we assess the advantages and limitations of metallodrug delivery, both from a nanocarrier and from a metal-nano interaction perspective. We describe the latest and most relevant nanomedicine formulations developed for metal complexes, and we discuss how the rational combination of coordination chemistry with nanomedicine technology can assist in promoting the clinical translation of metallodrugs.
Collapse
Affiliation(s)
- Quim Peña
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Alec Wang
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Orysia Zaremba
- BCMaterials, Bld. Martina Casiano, 3rd. Floor, UPV/EHU Science Park, 48940, Leioa, Spain
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Hans W Scheeren
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Josbert M Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Roger M Pallares
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Stefan Wuttke
- BCMaterials, Bld. Martina Casiano, 3rd. Floor, UPV/EHU Science Park, 48940, Leioa, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
31
|
Ravera M, Gabano E, McGlinchey MJ, Osella D. Pt(IV) antitumor prodrugs: dogmas, paradigms, and realities. Dalton Trans 2022; 51:2121-2134. [PMID: 35015025 DOI: 10.1039/d1dt03886a] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Platinum(II)-based drugs are widely used for the treatment of solid tumors, especially in combination protocols. Severe side effects and occurrence of resistance are the major limitations to their clinical use. To overcome these drawbacks, a plethora of Pt(IV) derivatives, acting as anticancer prodrugs, have been designed, synthesized and preclinically (often only in vitro) tested. Here, we summarize the recent progress in the development and understanding of the chemical properties and biochemical features of these Pt(IV) prodrugs, especially those containing bioactive molecules as axial ligands, acting as multi-functional agents. Even though no such prodrugs have been yet approved for clinical use, many show encouraging pharmacological profiles. Thus, a better understanding of their features is a promising approach towards improving the available Pt-based anticancer agents.
Collapse
Affiliation(s)
- Mauro Ravera
- Dipartimento di Scienze e Innovazione Tecnologica, Università del Piemonte Orientale, Viale Michel 11, Alessandria, Italy.
| | - Elisabetta Gabano
- Dipartimento di Scienze e Innovazione Tecnologica, Università del Piemonte Orientale, Viale Michel 11, Alessandria, Italy.
| | | | - Domenico Osella
- Dipartimento di Scienze e Innovazione Tecnologica, Università del Piemonte Orientale, Viale Michel 11, Alessandria, Italy.
| |
Collapse
|
32
|
Predarska I, Saoud M, Morgan I, Eichhorn T, Kaluđerović GN, Hey-Hawkins E. Cisplatin-cyclooxygenase inhibitor conjugates, free and immobilised in mesoporous silica SBA-15, prove highly potent against triple-negative MDA-MB-468 breast cancer cell line. Dalton Trans 2021; 51:857-869. [PMID: 34877948 DOI: 10.1039/d1dt03265h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
For the development of anticancer drugs with higher activity and reduced toxicity, two approaches were combined: preparation of platinum(IV) complexes exhibiting higher stability compared to their platinum(II) counterparts and loading them into mesoporous silica SBA-15 with the aim to utilise the passive enhanced permeability and retention (EPR) effect of nanoparticles for accumulation in tumour tissues. Three conjugates based on a cisplatin scaffold bearing the anti-inflammatory drugs naproxen, ibuprofen or flurbiprofen in the axial positions (1, 2 and 3, respectively) were synthesised and loaded into SBA-15 to afford the mesoporous silica nanoparticles (MSNs) SBA-15|1, SBA-15|2 and SBA-15|3. Superior antiproliferative activity of both free and immobilised conjugates in a panel of four breast cancer cell lines (MDA-MB-468, HCC1937, MCF-7 and BT-474) with markedly increased cytotoxicity with respect to cisplatin was demonstrated. All compounds exhibit highest activity against the triple-negative cell line MDA-MB-468, with conjugate 1 being the most potent. However, against MCF-7 and BT-474 cell lines, the most notable improvement was found, with IC50 values up to 240-fold lower than cisplatin. Flow cytometry assays clearly show that all compounds induce apoptotic cell death elevating the levels of both early and late apoptotic cells. Furthermore, autophagy as well as formation of reactive oxygen species (ROS) and nitric oxide (NO) were elevated to a similar or greater extent than with cisplatin.
Collapse
Affiliation(s)
- Ivana Predarska
- Universität Leipzig, Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Johannisallee 29, 04103 Leipzig, Germany. .,Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Mohamad Saoud
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany.
| | - Ibrahim Morgan
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany.
| | - Thomas Eichhorn
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Goran N Kaluđerović
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany.,Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany.
| | - Evamarie Hey-Hawkins
- Universität Leipzig, Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Johannisallee 29, 04103 Leipzig, Germany.
| |
Collapse
|