1
|
Florio D, Annunziata A, Panzetta V, Netti PA, Ruffo F, Marasco D. A Half-Sandwich Os(II) Glucoconjugated NHC Complex as a Modulator of Amyloid Aggregation. Inorg Chem 2025; 64:3335-3345. [PMID: 39945504 PMCID: PMC11863378 DOI: 10.1021/acs.inorgchem.4c04823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/25/2025]
Abstract
Herein, the effects of a novel half-sandwich Os(II) complex on the aggregation of an amyloid model system, derived from the C-terminal domain of the nucleophosmin 1 protein (NPM1264-277), were investigated. The thioflavin T (ThT) binding assay revealed that the complex [(η6-toluene)Os(NHCglu)Cl2] (where NHCglu is the N-heterocyclic carbene ligand 1-methyl-3-{2,3,4,6-tetra-O-acetyl-1-glucosyl}imidazol-2-ylidene), hence named Os-Tolu, was able to repress amyloid aggregation in a dose-dependent way. Conformational studies through circular dichroism (CD) and Fourier transform infrared (FTIR) spectroscopies clearly indicated that this inhibitory effect occurred through the stabilization of α-helical structures of monomeric NPM1264-277, thus hampering self-recognition. Electrospray ionization mass spectrometry (ESI-MS) studies evidenced, through the formation of coordination adducts, direct interactions of the amyloid peptide with the Os-glucoconjugate complex that, in turn, promote chemical modifications of the sequence further disfavoring the self-assembly process. Noticeably, the presence of Os-Tolu completely repressed the formation of amyloid fibers in scanning electron microscopy (SEM) analysis and induced a slight rescue of cell viability, in contrast to its reduction caused by the amyloid model in human SH-SY5Y neuroblastoma cells. These data strongly support the hypothesis of expanding the use of osmium-based agents to neurodegenerative diseases, positioning them as potential neurodrugs.
Collapse
Affiliation(s)
- Daniele Florio
- IRCCS SYNLAB
SDN, Via G., Ferraris
144, Naples 80146, Italy
| | - Alfonso Annunziata
- Department
of Chemical Sciences, University of Naples
Federico II, Naples 80126, Italy
| | - Valeria Panzetta
- Department
of Chemical, Materials, and Industrial Production Engineering (DICMaPI), University of Naples Federico II, Naples 80125, Italy
- Interdisciplinary
Research Centre on Biomaterials (CRIB), University of Naples Federico II, Istituto Italiano di Tecnologia, Naples 80125, Italy
| | - Paolo A. Netti
- Department
of Chemical, Materials, and Industrial Production Engineering (DICMaPI), University of Naples Federico II, Naples 80125, Italy
- Interdisciplinary
Research Centre on Biomaterials (CRIB), University of Naples Federico II, Istituto Italiano di Tecnologia, Naples 80125, Italy
| | - Francesco Ruffo
- Department
of Chemical Sciences, University of Naples
Federico II, Naples 80126, Italy
| | - Daniela Marasco
- Department
of Pharmacy, University of Naples Federico
II, Naples 80131, Italy
| |
Collapse
|
2
|
Guo X, Song J, Liu M, Ou X, Guo Y. The interplay between the tumor microenvironment and tumor-derived small extracellular vesicles in cancer development and therapeutic response. Cancer Biol Ther 2024; 25:2356831. [PMID: 38767879 PMCID: PMC11110713 DOI: 10.1080/15384047.2024.2356831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 05/14/2024] [Indexed: 05/22/2024] Open
Abstract
The tumor microenvironment (TME) plays an essential role in tumor cell survival by profoundly influencing their proliferation, metastasis, immune evasion, and resistance to treatment. Extracellular vesicles (EVs) are small particles released by all cell types and often reflect the state of their parental cells and modulate other cells' functions through the various cargo they transport. Tumor-derived small EVs (TDSEVs) can transport specific proteins, nucleic acids and lipids tailored to propagate tumor signals and establish a favorable TME. Thus, the TME's biological characteristics can affect TDSEV heterogeneity, and this interplay can amplify tumor growth, dissemination, and resistance to therapy. This review discusses the interplay between TME and TDSEVs based on their biological characteristics and summarizes strategies for targeting cancer cells. Additionally, it reviews the current issues and challenges in this field to offer fresh insights into comprehending tumor development mechanisms and exploring innovative clinical applications.
Collapse
Affiliation(s)
- Xuanyu Guo
- The Affiliated Hospital, Southwest Medical University, Luzhou, PR China
| | - Jiajun Song
- Department of Clinical Laboratory Medicine, the Affiliated Hospital, Southwest Medical University, Luzhou, PR China
| | - Miao Liu
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| | - Xinyi Ou
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| | - Yongcan Guo
- Nanobiosensing and Microfluidic Point-of-Care Testing, Key Laboratory of Luzhou, Department of Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, PR China
| |
Collapse
|
3
|
Jia Y, Wu Q, Yang Z, Sun R, Zhang K, Guo X, Xu R, Guo Y. Mechanisms of myocardial toxicity of antitumor drugs and potential therapeutic strategies: A review of the literature. Curr Probl Cardiol 2024; 49:102782. [PMID: 39134104 DOI: 10.1016/j.cpcardiol.2024.102782] [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: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
With the successive development of chemotherapy drugs, good results have been achieved in clinical application. However, myocardial toxicity is the biggest challenge. Anthracyclines, immune checkpoint inhibitors, and platinum drugs are widely used. Targeted drug delivery, nanomaterials and dynamic imaging evaluation are all emerging research directions. This article reviews the recent literature on the use of targeted nanodrug delivery and imaging techniques to evaluate the myocardial toxicity of antineoplastic drugs, and discusses the potential mechanisms.
Collapse
Affiliation(s)
- Yang Jia
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Qihong Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Zhigang Yang
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Street, Chengdu 610041, China
| | - Ran Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Kun Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Xia Guo
- Department of Hematology, West China Second University Hospital, Sichuan University; 20# South Renmin Road, Chengdu, Sichuan 610041, China
| | - Rong Xu
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China.
| | - Yingkun Guo
- Department of Radiology, West China Second University Hospital, Sichuan University, 20# South Renmin Road, Chengdu, Sichuan 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; 20# South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
4
|
Aher S, Zhu J, Bhagat P, Borse L, Liu X. Pt(IV) Complexes in the Search for Novel Platinum Prodrugs with Promising Activity. Top Curr Chem (Cham) 2024; 382:6. [PMID: 38400859 DOI: 10.1007/s41061-023-00448-3] [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: 07/14/2023] [Accepted: 11/15/2023] [Indexed: 02/26/2024]
Abstract
The kinetically inert, six coordinated, octahedral Pt(IV) complexes are termed dual-, triple-, or multi-action prodrugs based on the nature of the axially substituted ligands. These ligands are either inert or biologically active, where the nature of these axial ligands provides additional stability, synergistic biological activity or cell-targeting ability. There are many literature reports from each of these classes, mentioning the varied nature of these axial ligands. The ligands comprise drug molecules such as chlorambucil, doxorubicin, valproic acid, ethacrynic acid, biologically active chalcone, coumarin, combretastatin, non-steroidal anti-inflammatory drugs (NSAIDs) and many more, potentiating the anti-proliferative profile or reducing the side effects associated with cisplatin therapy. The targeting and non-targeting nature of these moieties exert additive or synergistic effects on the anti-cancer activity of Pt(II) moieties. Herein, we discuss the effects of these axially oriented ligands and the changes in the non-leaving am(m)ine groups and in the leaving groups on the biological activity. In this review, we have presented the latest developments in the field of Pt(IV) complexes that display promising activity with a reduced resistance profile. We have discussed the structure activity relationship (SAR) and the effects of the ligands on the biological activity of Pt(IV) complexes with cisplatin, oxaliplatin, carboplatin and the Pt core other than approved drugs. This literature work will help researchers to get an idea about Pt(IV) complexes that have been classified based on the aspects of their biological activity.
Collapse
Affiliation(s)
- Sainath Aher
- K. K. Wagh College of Pharmacy, Nashik, Maharashtra, 422003, India
- Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Molecular Sciences, Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, People's Republic of China
| | - Jinhua Zhu
- Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Molecular Sciences, Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, People's Republic of China
| | - Pundlik Bhagat
- Department of Chemistry, School of Advanced Sciences, VIT University, Vellore, 632014, India
| | - Laxmikant Borse
- Sandip Institute of Pharmaceutical Sciences, Nashik, Maharashtra, 422213, India
| | - Xiuhua Liu
- Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Molecular Sciences, Key Laboratory of Natural Medicine and Immuno-Engineering of Henan Province, Henan University, Kaifeng, 475004, People's Republic of China.
| |
Collapse
|
5
|
Farjadian F, Faghih Z, Fakhimi M, Iranpour P, Mohammadi-Samani S, Doroudian M. Glucosamine-Modified Mesoporous Silica-Coated Magnetic Nanoparticles: A "Raisin-Cake"-like Structure as an Efficient Theranostic Platform for Targeted Methotrexate Delivery. Pharmaceutics 2023; 15:2491. [PMID: 37896251 PMCID: PMC10610088 DOI: 10.3390/pharmaceutics15102491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
This study presents the synthesis of glucosamine-modified mesoporous silica-coated magnetic nanoparticles (MNPs) as a therapeutic platform for the delivery of an anticancer drug, methotrexate (MTX). The MNPs were coated with mesoporous silica in a templated sol-gel process to form MNP@MSN, and then chloropropyl groups were added to the structure in a post-modification reaction. Glucosamine was then reacted with the chloro-modified structure, and methotrexate was conjugated to the hydroxyl group of the glucose. The prepared structure was characterized using techniques such as Fourier transform infrared (FT-IR) spectroscopy, elemental analysis (CHN), field emission scanning electron microscopy (FESEM), transmission electron microscopy (TEM), dynamic light scattering (DLS), a vibrating sample magnetometer (VSM), and X-ray diffraction (XRD). Good formation of nano-sized MNPs and MNP@MSN was observed via particle size monitoring. The modified glucosamine structure showed a controlled release profile of methotrexate in simulated tumor fluid. In vitro evaluation using the 4T1 breast cancer cell line showed the cytotoxicity, apoptosis, and cell cycle effects of methotrexate. The MTT assay showed comparable toxicity between MTX-loaded nanoparticles and free MTX. The structure could act as a glucose transporter-targeting agent and showed increased uptake in cancer cells. An in vivo breast cancer model was established in BALB/C mice, and the distribution of MTX-conjugated MNP@MSN particles was visualized using MRI. The MTX-conjugated particles showed significant anti-tumor potential together with MRI contrast enhancement.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Canter, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran;
| | - Zahra Faghih
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45550, Iran; (Z.F.); (M.F.)
| | - Maryam Fakhimi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45550, Iran; (Z.F.); (M.F.)
| | - Pooya Iranpour
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz 71936-13311, Iran;
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Canter, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran;
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran
| |
Collapse
|
6
|
Pastuch-Gawołek G, Szreder J, Domińska M, Pielok M, Cichy P, Grymel M. A Small Sugar Molecule with Huge Potential in Targeted Cancer Therapy. Pharmaceutics 2023; 15:913. [PMID: 36986774 PMCID: PMC10056414 DOI: 10.3390/pharmaceutics15030913] [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: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The number of cancer-related diseases is still growing. Despite the availability of a large number of anticancer drugs, the ideal drug is still being sought that would be effective, selective, and overcome the effect of multidrug resistance. Therefore, researchers are still looking for ways to improve the properties of already-used chemotherapeutics. One of the possibilities is the development of targeted therapies. The use of prodrugs that release the bioactive substance only under the influence of factors characteristic of the tumor microenvironment makes it possible to deliver the drug precisely to the cancer cells. Obtaining such compounds is possible by coupling a therapeutic agent with a ligand targeting receptors, to which the attached ligand shows affinity and is overexpressed in cancer cells. Another way is to encapsulate the drug in a carrier that is stable in physiological conditions and sensitive to conditions of the tumor microenvironment. Such a carrier can be directed by attaching to it a ligand recognized by receptors typical of tumor cells. Sugars seem to be ideal ligands for obtaining prodrugs targeted at receptors overexpressed in cancer cells. They can also be ligands modifying polymers' drug carriers. Furthermore, polysaccharides can act as selective nanocarriers for numerous chemotherapeutics. The proof of this thesis is the huge number of papers devoted to their use for modification or targeted transport of anticancer compounds. In this work, selected examples of broad-defined sugars application for improving the properties of both already-used drugs and substances exhibiting anticancer activity are presented.
Collapse
Affiliation(s)
- Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Julia Szreder
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Monika Domińska
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Mateusz Pielok
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Piotr Cichy
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Mirosława Grymel
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| |
Collapse
|
7
|
Holzer I, Desiatkina O, Anghel N, Johns SK, Boubaker G, Hemphill A, Furrer J, Păunescu E. Synthesis and Antiparasitic Activity of New Trithiolato-Bridged Dinuclear Ruthenium(II)-arene-carbohydrate Conjugates. Molecules 2023; 28:902. [PMID: 36677958 PMCID: PMC9865825 DOI: 10.3390/molecules28020902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Eight novel carbohydrate-tethered trithiolato dinuclear ruthenium(II)-arene complexes were synthesized using CuAAC ‘click’ (Cu(I)-catalyzed azide-alkyne cycloaddition) reactions, and there in vitro activity against transgenic T. gondii tachyzoites constitutively expressing β-galactosidase (T. gondii β-gal) and in non-infected human foreskin fibroblasts, HFF, was determined at 0.1 and 1 µM. When evaluated at 1 µM, seven diruthenium-carbohydrate conjugates strongly impaired parasite proliferation by >90%, while HFF viability was retained at 50% or more, and they were further subjected to the half-maximal inhibitory concentration (IC50) measurement on T. gondii β-gal. Results revealed that the biological activity of the hybrids was influenced both by the nature of the carbohydrate (glucose vs. galactose) appended on ruthenium complex and the type/length of the linker between the two units. 23 and 26, two galactose-based diruthenium conjugates, exhibited low IC50 values and reduced effect on HFF viability when applied at 2.5 µM (23: IC50 = 0.032 µM/HFF viability 92% and 26: IC50 = 0.153 µM/HFF viability 97%). Remarkably, compounds 23 and 26 performed significantly better than the corresponding carbohydrate non-modified diruthenium complexes, showing that this type of conjugates are a promising approach for obtaining new antiparasitic compounds with reduced toxicity.
Collapse
Affiliation(s)
- Isabelle Holzer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Oksana Desiatkina
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Nicoleta Anghel
- Institute of Parasitology Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Serena K. Johns
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
- School of Chemistry, Cardiff University, Park Place, Cardiff CF103AT, UK
| | - Ghalia Boubaker
- Institute of Parasitology Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, 3012 Bern, Switzerland
| | - Julien Furrer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Emilia Păunescu
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| |
Collapse
|
8
|
Sharma N, Kabeer SW, Singh IP, Tikoo K. Cisplatin conjugation with an exopolysaccharide extracted from Lactobacillus gasseri potentiates its efficacy and attenuates its toxicity. Int J Biol Macromol 2023; 225:227-240. [PMID: 36354077 DOI: 10.1016/j.ijbiomac.2022.10.256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/05/2022]
Abstract
The development of newer cisplatin analogs is constantly being investigated owing to its low solubility, poor pharmacokinetics, and dose-related toxicity. In order to address the limitations of current cisplatin therapy, the present study was undertaken. Cisplatin conjugation with an exopolysaccharide extracted from Lactobacillus gasseri (LG-EPS) showed remarkably enhanced and selective anticancer activity by targeting tumor cells overexpressing glucose transporter 1 (GLUT1). The EPS-cisplatin complex exhibited a 600-fold increase in aqueous solubility with a better pharmacokinetic profile (longer half-life) in comparison to cisplatin. Cell viability assay and western blotting demonstrated a strong correlation between the cytotoxicity profile and GLUT1 expressions in different cell lines. The concentration of DNA-bound platinum was also found to be significantly higher in EPS-cisplatin-treated cells. Quercetin, a competitive inhibitor of GLUTs, was shown to prevent this selective uptake of EPS-cisplatin complex. Surprisingly, EPS-cisplatin complex showed an exceptionally safer profile (4 times the maximum tolerated dose of cisplatin) in the acute toxicity study and was also more efficacious against the xenograft mice model. The study suggests that this green glycoconjugation can be an effective and safer strategy to broaden the therapeutic potential of anti-cancer drugs in general and cisplatin in particular.
Collapse
Affiliation(s)
- Nisha Sharma
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - Shaheen Wasil Kabeer
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - Inder Pal Singh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar, Punjab 160062, India
| | - Kulbhushan Tikoo
- Laboratory of Epigenetics and Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
9
|
Shofolawe-Bakare OT, de Mel JU, Mishra SK, Hossain M, Hamadani CM, Pride MC, Dasanayake GS, Monroe W, Roth EW, Tanner EEL, Doerksen RJ, Smith AE, Werfel TA. ROS-Responsive Glycopolymeric Nanoparticles for Enhanced Drug Delivery to Macrophages. Macromol Biosci 2022; 22:e2200281. [PMID: 36125638 PMCID: PMC10013198 DOI: 10.1002/mabi.202200281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/12/1912] [Indexed: 02/02/2023]
Abstract
Macrophages play a diverse, key role in many pathologies, including inflammatory diseases, cardiovascular diseases, and cancer. However, many therapeutic strategies targeting macrophages suffer from systemic off-target toxicity resulting in notoriously narrow therapeutic windows. To address this shortcoming, the development of poly(propylene sulfide)-b-poly(methacrylamidoglucopyranose) [PPS-b-PMAG] diblock copolymer-based nanoparticles (PMAG NPs) capable of targeting macrophages and releasing drug in the presence of reactive oxygen species (ROS) is reported. PMAG NPs have desirable physicochemical properties for systemic drug delivery, including slightly negative surface charge, ≈100 nm diameter, and hemo-compatibility. Additionally, due to the presence of PPS in the NP core, PMAG NPs release drug cargo preferentially in the presence of ROS. Importantly, PMAG NPs display high cytocompatibility and are taken up by macrophages in cell culture at a rate ≈18-fold higher than PEGMA NPs-NPs composed of PPS-b-poly(oligoethylene glycol methacrylate). Computational studies indicate that PMAG NPs likely bind with glucose transporters such as GLUT 1/3 on the macrophage cell surface to facilitate high levels of internalization. Collectively, this study introduces glycopolymeric NPs that are uniquely capable of both receptor-ligand targeting to macrophages and ROS-dependent drug release and that can be useful in many immunotherapeutic settings.
Collapse
Affiliation(s)
| | - Judith U de Mel
- Department of Biomedical Engineering, University of Mississippi, University, MS, 38677, USA
| | - Sushil K Mishra
- Department of BioMolecular Sciences, University of Mississippi, University, MS, 38677, USA
| | - Mehjabeen Hossain
- Department of BioMolecular Sciences, University of Mississippi, University, MS, 38677, USA
| | - Christine M Hamadani
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, 38677, USA
| | - Mercedes C Pride
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, 38677, USA
| | - Gaya S Dasanayake
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, 38677, USA
| | - Wake Monroe
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, 38677, USA
| | - Eric W Roth
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Eden E L Tanner
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, 38677, USA
| | - Robert J Doerksen
- Department of BioMolecular Sciences, University of Mississippi, University, MS, 38677, USA
| | - Adam E Smith
- Department of Chemical Engineering, University of Mississippi, University, MS, 38677, USA
- Department of Biomedical Engineering, University of Mississippi, University, MS, 38677, USA
| | - Thomas A Werfel
- Department of Chemical Engineering, University of Mississippi, University, MS, 38677, USA
- Department of Biomedical Engineering, University of Mississippi, University, MS, 38677, USA
- Department of BioMolecular Sciences, University of Mississippi, University, MS, 38677, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| |
Collapse
|
10
|
Jia C, Deacon GB, Zhang Y, Gao C. Platinum(IV) antitumor complexes and their nano-drug delivery. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213640] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
11
|
Fu J, Yang J, Seeberger PH, Yin J. Glycoconjugates for glucose transporter-mediated cancer-specific targeting and treatment. Carbohydr Res 2020; 498:108195. [PMID: 33220603 DOI: 10.1016/j.carres.2020.108195] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/29/2022]
Abstract
First observed in 1920s, the Warburg effects have inspired scientists to harness the unique glucose metabolism of cancer cells for targeted therapy for a century. Carbohydrate-drug conjugates are explicitly designed for selective uptake by cancer cells overexpressing glucose transporters. We summarize the progress in developing glycoconjugates for cancer-specific targeting and treatment over the past decade (2010-2020) and point to some future directions in this field.
Collapse
Affiliation(s)
- Junjie Fu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, PR China
| | - Jiaxin Yang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, PR China
| | - Peter H Seeberger
- Biomolecular Systems Department, Max Planck Institute of Colloids and Interfaces, Potsdam, 14476, Germany
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, PR China.
| |
Collapse
|
12
|
Bononi G, Iacopini D, Cicio G, Di Pietro S, Granchi C, Di Bussolo V, Minutolo F. Glycoconjugated Metal Complexes as Cancer Diagnostic and Therapeutic Agents. ChemMedChem 2020; 16:30-64. [PMID: 32735702 DOI: 10.1002/cmdc.202000456] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Indexed: 12/15/2022]
Abstract
The possibility of selectively delivering metal complexes to a defined cohort of cells on the basis of their metabolic features is a highly challenging goal, which may be extremely useful for a series of purposes, including diagnosis and therapy of pathological states, such as cancer. Tumor cells display augmented requests for carbohydrates and, in particular, for glucose in order to sustain their high proliferation rate, which causes an increased glycolytic process (Warburg effect). Since several metal complexes display diagnostic and/or therapeutic properties, their conjugation to carbohydrate portions often induce their preferential accumulation in cancer cells, similarly to what is observed with fluorodeoxyglucose (FDG). In this review we have considered the latest developments of glycoconjugates containing metal complexes in their structures. These compounds are classified as diagnostic or therapeutic agents and are further systematically discussed on the basis of the metal atom they contain. Several diagnostic techniques are possible with these probes, since, depending on the metal species included in their structures, they may be employed in nuclear medicine (PET, SPECT), magnetic resonance imaging, luminescence and phosphorescence. At the same time, the lack of selective cytotoxicity displayed by several metal-based chemotherapeutic agents, may also be solved by the conjugation of these agents to carbohydrate portions. Overall, data so far available reveal the great potential of this chemical class in the early detection and in the cure of severe neoplastic diseases, which still needs to be fully explored in the clinic.
Collapse
Affiliation(s)
- Giulia Bononi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno Pisano 33, 56126, Pisa, Italy
| | - Dalila Iacopini
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi 13, 56124, Pisa, Italy
| | - Gaspare Cicio
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno Pisano 33, 56126, Pisa, Italy.,Current address: Menarini Ricerche S.p.A. -, Laboratori di Pisa, Via Livornese 897, 56122, Pisa, Italy
| | - Sebastiano Di Pietro
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno Pisano 33, 56126, Pisa, Italy
| | - Carlotta Granchi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno Pisano 33, 56126, Pisa, Italy
| | - Valeria Di Bussolo
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi 13, 56124, Pisa, Italy
| | - Filippo Minutolo
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno Pisano 33, 56126, Pisa, Italy
| |
Collapse
|
13
|
Khoury A, Deo KM, Aldrich-Wright JR. Recent advances in platinum-based chemotherapeutics that exhibit inhibitory and targeted mechanisms of action. J Inorg Biochem 2020; 207:111070. [PMID: 32299045 DOI: 10.1016/j.jinorgbio.2020.111070] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/22/2022]
Abstract
Current platinum-based drugs used in chemotherapy, like cisplatin and its derivatives, are greatly limited due to side-effects and drug resistance. This has inspired the search for novel platinum-based drugs that deviate from the conventional mechanism of action seen with current chemotherapeutics. This review highlights recent advances in platinum(II) and platinum(IV)-based complexes that have been developed within the past six years. The platinum compounds explored within this review are those that display a more targeted approach by incorporating ligands that act on selected cellular targets within cancer cells. This includes mitochondria, overexpressed receptors or proteins and enzymes that contribute to cancer cell proliferation. These types of platinum compounds have shown significant improvements in anticancer activity and as such, this review highlights the importance of pursuing these new designed platinum drugs for cancer therapy, with the potential of undergoing clinical trials.
Collapse
Affiliation(s)
- Aleen Khoury
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Krishant M Deo
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | | |
Collapse
|
14
|
Annunziata A, Amoresano A, Cucciolito ME, Esposito R, Ferraro G, Iacobucci I, Imbimbo P, Lucignano R, Melchiorre M, Monti M, Scognamiglio C, Tuzi A, Monti DM, Merlino A, Ruffo F. Pt(II) versus Pt(IV) in Carbene Glycoconjugate Antitumor Agents: Minimal Structural Variations and Great Performance Changes. Inorg Chem 2020; 59:4002-4014. [PMID: 32129608 PMCID: PMC7997382 DOI: 10.1021/acs.inorgchem.9b03683] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Indexed: 12/20/2022]
Abstract
Octahedral Pt(IV) complexes (2Pt-R) containing a glycoconjugate carbene ligand were prepared and fully characterized. These complexes are structural analogues to the trigonal bipyramidal Pt(II) species (1Pt-R) recently described. Thus, an unprecedented direct comparison between the biological properties of Pt compounds with different oxidation states and almost indistinguishable structural features was performed. The stability profile of the novel Pt(IV) compounds in reference solvents was determined and compared to that of the analogous Pt(II) complexes. The uptake and antiproliferative activities of 2Pt-R and 1Pt-R were evaluated on the same panel of cell lines. DNA and protein binding properties were assessed using human serum albumin, the model protein hen egg white lysozyme, and double stranded DNA model systems by a variety of experimental techniques, including UV-vis absorption spectroscopy, fluorescence, circular dichroism, and electrospray ionization mass spectrometry. Although the compounds present similar structures, their in-solution stability, cellular uptake, and DNA binding properties are diverse. These differences may represent the basis of their different cytotoxicity and biological activity.
Collapse
Affiliation(s)
- Alfonso Annunziata
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Angela Amoresano
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Maria Elena Cucciolito
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
- CIRCC, via Celso Ulpiani
27, 70126 Bari, Italy
| | - Roberto Esposito
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
- CIRCC, via Celso Ulpiani
27, 70126 Bari, Italy
| | - Giarita Ferraro
- Dipartimento di Chimica Ugo Schiff, Università di Firenze, Sesto Fiorentino, Florence 50019, Italy
| | - Ilaria Iacobucci
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Paola Imbimbo
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Rosanna Lucignano
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | | | - Maria Monti
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Chiara Scognamiglio
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Angela Tuzi
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Daria Maria Monti
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Antonello Merlino
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
| | - Francesco Ruffo
- Dipartimento di
Scienze Chimiche, Università di Napoli
Federico II, Complesso Universitario di Monte S. Angelo, via Cintia 21, 80126 Napoli, Italy
- CIRCC, via Celso Ulpiani
27, 70126 Bari, Italy
| |
Collapse
|
15
|
Liu H, Ma J, Li Y, Yue K, Li L, Xi Z, Zhang X, Liu J, Feng K, Ma Q, Liu S, Guo S, Wang PG, Wang C, Xie S. Polyamine-Based Pt(IV) Prodrugs as Substrates for Polyamine Transporters Preferentially Accumulate in Cancer Metastases as DNA and Polyamine Metabolism Dual-Targeted Antimetastatic Agents. J Med Chem 2019; 62:11324-11334. [PMID: 31765154 DOI: 10.1021/acs.jmedchem.9b01641] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diverse platinum drug candidates have been designed to improve inhibitory potency and overcome resistance for orthotopic tumors. However, the antimetastatic properties have rarely been reported. We herein report that homospermidineplatin (4a), a polyamine-Pt(IV) prodrug, can potently inhibit tumor growth in situ and reverse cisplatin resistance as expected, and more importantly, 4a displays remarkably elevated antimetastatic activity in vivo (65.7%), compared to those of cisplatin (27.0%) and oxaliplatin (19.6%). The underlying molecular mechanism indicates that in addition to targeting nuclear DNA, 4a can modulate polyamine metabolism and function in a manner different from that of cisplatin. By upregulating SSAT and PAO, 4a downregulates the concentrations of Put, Spd, and Spm, which favors the suppression of fast-growing tumor cells. Moreover, the p53/SSAT/β-catenin and PAO/ROS/GSH/GSH-Px pathways are involved in the inhibition of 4a-induced tumor metastasis. Our study implies a promising strategy for the design of platinum drugs for the treatment of terminal cancer.
Collapse
Affiliation(s)
- Hanfang Liu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Jing Ma
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Yingguang Li
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Kexin Yue
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Linrong Li
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Zhuoqing Xi
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
- Henan University of Science and Technology Second Affiliated Hospital , Luoyang 471000 , China
| | - Xiao Zhang
- The Key Laboratory of Natural Medicine and Immuno-Engineering , Henan University , Kaifeng 475004 , China
| | - Jianing Liu
- School of Medicine , Henan University Minsheng College , Kaifeng 475004 , China
| | - Kai Feng
- School of Medicine , Henan University Minsheng College , Kaifeng 475004 , China
| | - Qi Ma
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Sitong Liu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Shudi Guo
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| | - Peng George Wang
- The State Key Laboratory of Microbial Technology and National Glycoengineering Research Center , Shandong University , Qingdao 266237 , China
| | - Chaojie Wang
- The Key Laboratory of Natural Medicine and Immuno-Engineering , Henan University , Kaifeng 475004 , China
| | - Songqiang Xie
- School of Pharmacy, Institute of Chemical Biology , Henan University , North Jinming Avenue , Kaifeng 475004 , China
| |
Collapse
|