1
|
Su J, Yan T, Zhang X, Yan T, Wang Z. Tumor microenvironment-responsive diagnosis and treatment of integrated drug-loaded CdTe quantum dots for treatment tumors. NANOTECHNOLOGY 2025; 36:235101. [PMID: 40345213 DOI: 10.1088/1361-6528/add6ad] [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: 11/19/2024] [Accepted: 05/09/2025] [Indexed: 05/11/2025]
Abstract
The acidic tumor microenvironment is a common feature of tumors, and boric acid-functionalized quantum dots (BA-QDs) exhibit pH-sensitive boron affinity effects and fluorescence emission characteristics. In this study, CdTe QDs were prepared using the water phase synthesis method. Additionally, BA-QDs were prepared by modifying QDs with 4-mercaptophenylboric acid. Hesperetin, baicalein, quercetin, and other model drugs were used, with QDs and BA-QDs serving as carriers, to create a drug-loaded system of QDs with tumor microenvironment-responsive drug release performance. The physical and chemical properties were characterized using dynamic light scattering (DLS), Fourier transform infrared spectroscopy, transmission electron microscopy, x-ray diffraction, etc. Our findings showed that the synthesis of drug-loaded QDs with a uniform particle size was successful. The experiments involved studying the adsorption kinetics of the QDs and the degree of dissolution of the drug-loaded QDsin vitro. BA-QDs exhibited pH-responsive drug release and fluorescence emission properties.In vitrocell experiments were conducted to examine the uptake and imaging effects of QDs and BA-QDs at the cell level. The results showed that both QDs and BA-QDs exhibited effective imaging at the cell level. Moreover, the three drug-loaded BA-QDs inhibited HepG2 cancer cells by about 80%, indicating a significant inhibitory effect on cancer cells. Here, we developed a universal new technology for tumor diagnosis and treatment, provided innovative approaches for targeted cancer diagnosis and treatment, and broadened the application scope of nanofluorescence technology.
Collapse
Affiliation(s)
- Jiahao Su
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, People's Republic of China
| | - Tingyuan Yan
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, Jiangsu, People's Republic of China
| | - Xiankang Zhang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, People's Republic of China
| | - Tingxuan Yan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243032, People's Republic of China
| | - Zhixiang Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, Jiangsu, People's Republic of China
| |
Collapse
|
2
|
Yang L, Wu W, Yang J, Xu M. Nanoparticle-mediated delivery of herbal-derived natural products to modulate immunosenescence-induced drug resistance in cancer therapy: a comprehensive review. Front Oncol 2025; 15:1567896. [PMID: 40356750 PMCID: PMC12066338 DOI: 10.3389/fonc.2025.1567896] [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] [Received: 01/28/2025] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Immunosenescence, the age-associated decline of the immune system, is pivotal in fostering drug resistance within the tumor microenvironment (TME). The accumulation of senescent immune cells and the release of pro-inflammatory senescence-associated secretory phenotype (SASP) factors create a milieu that supports tumor survival and undermines therapeutic efficacy. Traditional cancer treatments often fail to address this underlying issue, leading to suboptimal outcomes. This article proposes an innovative strategy to overcome immunosenescence-induced drug resistance through the nanoparticle-mediated delivery of herbal-derived natural products (HDNPs), which possess senolytic and immunomodulatory properties capable of clearing senescent cells and rejuvenating immune function. Nanoparticle delivery systems enhance these compounds' stability, bioavailability, and targeted delivery to the TME and senescent immune cells. By harnessing the synergistic effects of HDNPs and nanotechnology, this approach offers a novel and multifaceted solution to drug resistance in cancer therapy. It holds the potential to restore immune surveillance, reduce pro-survival signaling in cancer cells, and enhance the efficacy of conventional treatments. This paradigm shift emphasizes the importance of addressing immunosenescence as a therapeutic target and paves the way for more effective and personalized cancer interventions.
Collapse
Affiliation(s)
- Lichang Yang
- Xuzhou Affiliated Hospital of Nanjing University of Chinese Medicine, Xuzhou, China
| | - Wei Wu
- Department of Geriatrics, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Yang
- Xuzhou Affiliated Hospital of Nanjing University of Chinese Medicine, Xuzhou, China
| | - Manman Xu
- Department of Geriatrics, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Ahmed ZSO, Khan E, Elias N, Elshebiny A, Dou Q. Updated Review on Natural Polyphenols: Molecular Mechanisms, Biological Effects, and Clinical Applications for Cancer Management. Biomolecules 2025; 15:629. [PMID: 40427522 PMCID: PMC12108987 DOI: 10.3390/biom15050629] [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: 03/10/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Polyphenols, naturally occurring compounds found exclusively in plants, have gained significant attention for their potential in cancer prevention and treatment. These compounds are known for their antioxidant properties and are abundant in various plant-based foods, such as vegetables, fruits, grains, and beverages. Recent studies have highlighted the broad spectrum of health benefits of polyphenols, including their antiviral, anti-inflammatory, and anticancer properties. In addition, these naturally derived compounds are increasingly important for drug discovery due to their high molecular diversity and novel biofunctionalities. This review provides an in-depth analysis of the current research and knowledge on the potential use of dietary polyphenols as bioactive compounds for the prevention and treatment of various cancers. This review aims to provide valuable insights into the mechanisms underlying the anticancer properties of phenolic compounds in both laboratory and clinical settings. Furthermore, this review highlights the positive clinical outcomes associated with the use of polyphenols as anticancer agents and offers guidance for future research to advance this promising field.
Collapse
Affiliation(s)
- Zainab Sabry Othman Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, King Salman International University, Ras Sudr 46612, Egypt
| | - Elyas Khan
- Departments of Oncology, Pharmacology and Pathology School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA; (E.K.); (N.E.); (A.E.)
| | - Nathan Elias
- Departments of Oncology, Pharmacology and Pathology School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA; (E.K.); (N.E.); (A.E.)
| | - Alhussein Elshebiny
- Departments of Oncology, Pharmacology and Pathology School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA; (E.K.); (N.E.); (A.E.)
| | - Qingping Dou
- Departments of Oncology, Pharmacology and Pathology School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA; (E.K.); (N.E.); (A.E.)
| |
Collapse
|
4
|
Uti DE, Atangwho IJ, Alum EU, Ntaobeten E, Obeten UN, Bawa I, Agada SA, Ukam CIO, Egbung GE. Antioxidants in cancer therapy mitigating lipid peroxidation without compromising treatment through nanotechnology. DISCOVER NANO 2025; 20:70. [PMID: 40272665 PMCID: PMC12021792 DOI: 10.1186/s11671-025-04248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/27/2025]
Abstract
BACKGROUND Cancer treatments often exploit oxidative stress to selectively kill tumour cells by disrupting their lipid peroxidation membranes and inhibiting antioxidant enzymes. However, lipid peroxidation plays a dual role in cancer progression, acting as both a tumour promoter and a suppressor. Balancing oxidative stress through antioxidant therapy remains a challenge, as excessive antioxidant activity may compromise the efficacy of chemotherapy and radiotherapy. AIM This review explores the role of antioxidants in mitigating lipid peroxidation in cancer therapy while maintaining treatment efficacy. It highlights recent advancements in nanotechnology-based targeted antioxidant delivery to optimize therapeutic outcomes. METHODS A comprehensive literature review was conducted using reputable databases, including PubMed, Scopus, Web of Science, and ScienceDirect. The search focused on publications from the past five years (2020-2025), supplemented by relevant studies from earlier years. Keywords such as "antioxidants," "lipid peroxidation," "nanotechnology in cancer therapy," and "oxidative stress" were utilized. Relevant articles were critically analysed, and graphical illustrations were created. RESULTS Emerging evidence suggests that nanoparticles, including liposomes, polymeric nanoparticles, metal-organic frameworks, and others, can effectively encapsulate and control the release of antioxidants in tumour cells while minimizing systemic toxicity. Stimuli-responsive carriers with tumour-specific targeting mechanisms further enhance antioxidant delivery. Studies indicate that these strategies help preserve normal cells, mitigate oxidative stress-related damage, and improve treatment efficacy. However, challenges such as bioavailability, stability, and potential interactions with standard therapies remain. CONCLUSION Integrating nanotechnology with antioxidant-based interventions presents a promising approach for optimizing cancer therapy. Future research should focus on refining lipid peroxidation modulation strategies, assessing oxidative stress profiles during treatment, and employing biomarkers to determine optimal antioxidant dosing. A balanced approach to antioxidant use may enhance therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
- Daniel Ejim Uti
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria.
| | - Item Justin Atangwho
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Esther Ugo Alum
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda
| | - Emmanuella Ntaobeten
- Department of Cancer and Haematology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Uket Nta Obeten
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| | - Inalegwu Bawa
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | - Samuel A Agada
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | | | - Godwin Eneji Egbung
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
5
|
Teimouri M, Homayouni Tabrizi M, Karimi E. The selective proapoptotic impact of the myricetin-loaded alginate-cellulose hybrid nanocrystals (MAC-NCs) on the human AGS gastric cancer cells. Mol Biol Rep 2024; 51:998. [PMID: 39299971 DOI: 10.1007/s11033-024-09864-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/14/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Myricetin, a flavanol present in fruits, tea, and vegetables, has the potential to reduce chronic diseases like gastric cancer by promoting cell death and stopping cell growth. However, its limited bioactivity due to its short lifespan and poor solubility in water has been a challenge. The current research focuses on incorporating myricetin into alginate-cellulose hybrid nanocrystals to enhance its selective proapoptotic effects on human AGS gastric cancer cells. METHODS MAC-NCs, myricetin-loaded alginate-cellulose hybrid nanocrystals, were synthesized using a combined co-precipitation/ultrasonic homogenization method and characterized through Dynamic Light Scattering (DLS), Fourier Transform Infrared Spectroscopy (FTIR), Field Emission Scanning Electron Microscope (FESEM), and Zeta-potential analyses. Their cytotoxic activity was tested on cancerous (AGS) and normal (Huvec) cells, revealing selective toxicity. Apoptotic markers, Caspase 8 and Caspase 9, gene expression was measured, and cell death type was confirmed using DAPI staining and flow cytometry on AGS cells. RESULTS Synthesized MAC-NCs, measuring 40 nm, showed significant selective toxicity on human gastric cells (IC50 of 31.05 µg/mL) compared to normal endothelial cells (IC50 of 214.26 µg/mL). DAPI and annexin flow cytometry revealed increased apoptotic bodies in gastric cells, indicating apoptosis. However, the apoptosis was found to be independent of Caspase-8 and Caspase-9. CONCLUSION The current study provides critical insights into the therapeutic potential of MAC-NCs for gastric cancer treatment. Based on the notable induction of apoptosis in the AGS cancer cell line, the synthesized MAC-NCs exhibit promising potential as a selective anti-gastric cancer agent. However, further in-vivo studies are necessary to confirm and quantify the nanoparticle's selective toxicity and pharmaceutical properties in future investigations.
Collapse
Affiliation(s)
- Mahdieh Teimouri
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Ehsan Karimi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
6
|
Samantaray A, Pradhan D, Nayak NR, Chawla S, Behera B, Mohanty L, Bisoyi SK, Gandhi S. Nanoquercetin based nanoformulations for triple negative breast cancer therapy and its role in overcoming drug resistance. Discov Oncol 2024; 15:452. [PMID: 39287822 PMCID: PMC11408462 DOI: 10.1007/s12672-024-01239-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Triple Negative Breast Cancer (TNBC) is a highly aggressive and treatment-resistant subtype of breast cancer, lacking the expression of estrogen, progesterone, and HER2 receptors. Conventional chemotherapy remains the primary treatment option, but its efficacy is often compromised by the development of drug resistance. Nanoquercetin has garnered the attention of researchers due to its potential in combating cancer. This antioxidant exhibits significant efficacy against various types of cancer, including blood, breast, pancreatic, prostate, colon, and oral cancers. Functioning as a potential anti-cancer agent, nanoquercetin impedes the development and proliferation of cancer cells, induces apoptosis and autophagy, and prevents cancer cell invasion and metastasis. Numerous processes, such as the inhibition of pathways linked to angiogenesis, inflammation, and cell survival, are responsible for these anticancer actions. Moreover, it shields DNA from degradation caused by radiation and other carcinogens. The cost-effectiveness of current cancer treatments remains a significant challenge in healthcare, imposing a substantial economic burden on societies worldwide. Preclinical studies and early-phase clinical trials indicate that nanoquercetin-based therapies could offer a significant advancement in the management of TNBC, providing a foundation for future research and clinical application in overcoming drug resistance and improving patient outcomes. This article examines the latest data on nanoquercetin's potent anti-cancer properties and interprets the accumulated research findings within the framework of preventive, predictive, and personalized (3P) medicine.
Collapse
Affiliation(s)
- Adyasa Samantaray
- University Department of Pharmaceutical Sciences, Utkal University, Vani Vihar, Bhubaneswar, Odisha, India
| | - Debasish Pradhan
- University Department of Pharmaceutical Sciences, Utkal University, Vani Vihar, Bhubaneswar, Odisha, India.
| | - Nalini Ranjan Nayak
- University Department of Pharmaceutical Sciences, Utkal University, Vani Vihar, Bhubaneswar, Odisha, India
| | - Saurabh Chawla
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, Khurda, Odisha, India
| | - Bandana Behera
- Faculty of Pharmacy, C.V.Raman Global University, Bhubaneswar, India
| | - Lalatendu Mohanty
- Department of Pharmaceutical Sciences, HNB Garhwal University, Uttarakhand, India
| | - Saroj Kanta Bisoyi
- University Department of Pharmaceutical Sciences, Utkal University, Vani Vihar, Bhubaneswar, Odisha, India
| | - Sabnam Gandhi
- University Department of Pharmaceutical Sciences, Utkal University, Vani Vihar, Bhubaneswar, Odisha, India
| |
Collapse
|
7
|
Elbeltagi S, Abdel Shakor AB, M Alharbi H, Tawfeek HM, Aldosari BN, E Eldin Z, Amin BH, Abd El-Aal M. Synergistic effects of quercetin-loaded CoFe 2O 4@Liposomes regulate DNA damage and apoptosis in MCF-7 cancer cells: based on biophysical magnetic hyperthermia. Drug Dev Ind Pharm 2024; 50:561-575. [PMID: 38832870 DOI: 10.1080/03639045.2024.2363231] [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: 04/22/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION Breast cancer (BC) is the most common malignancy in women globally. Significant progress has been made in developing structural nanoparticles (NPs) and formulations for targeted smart drug delivery (SDD) of pharmaceuticals, improving the precision of tumor cell targeting in therapy. SIGNIFICANCE Magnetic hyperthermia (MHT) treatment using magneto-liposomes (MLs) has emerged as a promising adjuvant cancer therapy. METHODS CoFe2O4 magnetic NPs (MNPs) were conjugated with nanoliposomes to form MLs, and the anticancer drug quercetin (Que) was loaded into MLs, forming Que-MLs composites for antitumor approach. The aim was to prepare Que-MLs for DD systems (DDS) under an alternating magnetic field (AMF), termed chemotherapy/hyperthermia (chemo-HT) techniques. The encapsulation efficiency (EE), drug loading capacity (DL), and drug release (DR) of Que and Que-MLs were evaluated. RESULTS The results confirmed successful Que-loading on the surface of MLs, with an average diameter of 38 nm and efficient encapsulation into MLs (69%). In vitro, experimental results on MCF-7 breast cells using MHT showed high cytotoxic effects of novel Que-MLs on MCF-7 cells. Various analyses, including cytotoxicity, apoptosis, cell migration, western blotting, fluorescence imaging, and cell membrane internalization, were conducted. The Acridine Orange-ethidium bromide double fluorescence test identified 35% early and 55% late apoptosis resulting from Que-MLs under the chemo-HT group. TEM results indicated MCF-7 cell membrane internalization and digestion of Que-MLs, suggesting the presence of early endosome-like vesicles on the cytoplasmic periphery. CONCLUSIONS Que-MLs exhibited multi-modal chemo-HT effects, displaying high toxicity against MCF-7 BC cells and showing promise as a potent cytotoxic agent for BC chemotherapy.
Collapse
Affiliation(s)
- Shehab Elbeltagi
- Department of Physics-Biophysics, Faculty of Science, New Valley University, New Valley, Egypt
| | - Abo Bakr Abdel Shakor
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
- School of biotechnology, Badr University in Assiut (BUA), Egypt
| | - Hanan M Alharbi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Hesham M Tawfeek
- Industrial Pharmacy Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Basmah N Aldosari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Zienab E Eldin
- Department of Material Science and nanotechnology, (PSAS), Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
- Center for Material Science, Zewail City of Science and Technology, Giza, Egypt
| | - Basma H Amin
- The Regional Center for Mycology and Biotechnology (RCMB), Al - Azhar University, Egypt
| | - Mohamed Abd El-Aal
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| |
Collapse
|
8
|
Caro C, Guzzi C, Moral-Sánchez I, Urbano-Gámez JD, Beltrán AM, García-Martín ML. Smart Design of ZnFe and ZnFe@Fe Nanoparticles for MRI-Tracked Magnetic Hyperthermia Therapy: Challenging Classical Theories of Nanoparticles Growth and Nanomagnetism. Adv Healthc Mater 2024; 13:e2304044. [PMID: 38303644 DOI: 10.1002/adhm.202304044] [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: 11/17/2023] [Revised: 01/18/2024] [Indexed: 02/03/2024]
Abstract
Iron Oxide Nanoparticles (IONPs) hold the potential to exert significant influence on fighting cancer through their theranostics capabilities as contrast agents (CAs) for magnetic resonance imaging (MRI) and as mediators for magnetic hyperthermia (MH). In addition, these capabilities can be improved by doping IONPs with other elements. In this work, the synthesis and characterization of single-core and alloy ZnFe novel magnetic nanoparticles (MNPs), with improved magnetic properties and more efficient magnetic-to-heat conversion, are reported. Remarkably, the results challenge classical nucleation and growth theories, which cannot fully predict the final size/shape of these nanoparticles and, consequently, their magnetic properties, implying the need for further studies to better understand the nanomagnetism phenomenon. On the other hand, leveraging the enhanced properties of these new NPs, successful tumor therapy by MH is achieved following their intravenous administration and tumor accumulation via the enhanced permeability and retention (EPR) effect. Notably, these results are obtained using a single low dose of MNPs and a single exposure to clinically suitable alternating magnetic fields (AMF). Therefore, as far as the authors are aware, for the first time, the successful application of intravenously administered MNPs for MRI-tracked MH tumor therapy in passively targeted tumor xenografts using clinically suitable conditions is demonstrated.
Collapse
Affiliation(s)
- Carlos Caro
- Biomedical Magnetic Resonance Laboratory-BMRL, Andalusian Public Foundation Progress and Health-FPS, Seville, 41092, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Universidad de Málaga, C/Severo Ochoa, 35, Malaga, 29590, Spain
| | - Cinzia Guzzi
- Biomedical Magnetic Resonance Laboratory-BMRL, Andalusian Public Foundation Progress and Health-FPS, Seville, 41092, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Universidad de Málaga, C/Severo Ochoa, 35, Malaga, 29590, Spain
| | - Irene Moral-Sánchez
- Biomedical Magnetic Resonance Laboratory-BMRL, Andalusian Public Foundation Progress and Health-FPS, Seville, 41092, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Universidad de Málaga, C/Severo Ochoa, 35, Malaga, 29590, Spain
| | - Jesús David Urbano-Gámez
- Biomedical Magnetic Resonance Laboratory-BMRL, Andalusian Public Foundation Progress and Health-FPS, Seville, 41092, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Universidad de Málaga, C/Severo Ochoa, 35, Malaga, 29590, Spain
| | - Ana M Beltrán
- Departamento de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, Universidad de Sevilla, Virgen de África 7, Sevilla, 41011, Spain
| | - Maria Luisa García-Martín
- Biomedical Magnetic Resonance Laboratory-BMRL, Andalusian Public Foundation Progress and Health-FPS, Seville, 41092, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Universidad de Málaga, C/Severo Ochoa, 35, Malaga, 29590, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Madrid, 28029, Spain
| |
Collapse
|
9
|
Ostovar S, Pourmadadi M, Zaker MA. Co-biopolymer of chitosan/carboxymethyl cellulose hydrogel improved by zinc oxide and graphene quantum dots nanoparticles as pH-sensitive nanocomposite for quercetin delivery to brain cancer treatment. Int J Biol Macromol 2023; 253:127091. [PMID: 37758113 DOI: 10.1016/j.ijbiomac.2023.127091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/16/2023] [Accepted: 09/24/2023] [Indexed: 10/02/2023]
Abstract
Brain cancer is the major reason of cancer-relevant deaths every year, as it is the most challenging cancer to treat and drug delivery. Quercetin (QUR), as a flavonoid substance found in plants and fruits, has good anticancer and medicinal effects on brain tumors, but its low stability and bioavailability as well as the blood-brain barrier (BBB), prevent it from reaching brain tumors. This research has introduced a nanocomposite made of biocompatible polymers, chitosan, and carboxymethyl cellulose. This co- biopolymer's mechanical and chemical properties and drug-loading capacity have been improved by adding zinc oxide nanoparticles (ZnO NPs). In addition, graphene quantum dots (GQDs) were used to improve the chemical properties as well as the ability to penetrate the BBB. The CS/CMC/GQDs/ZnO@QUR nanocomposites have nanoneedle structures with an average size of 219.38 ± 5.21 nm and a zeta potential of -53 mV. The morphology, chemical bonds, and crystallinity of the nanocomposite were examined by FE-SEM, FTIR, and XRD analyses, respectively. By examining the release of QUR, it became apparent that the half-drug release takes about 72 h, which has a much more controlled release than other QUR carriers. Further, the MTT test on U-87 MG and L929 cell lines suggested that this nanocomposite has good anticancer properties and low cytotoxicity compared to the free QUR.
Collapse
Affiliation(s)
- Shima Ostovar
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4563, Iran
| | - Mehrab Pourmadadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 11155-4563, Iran.
| | - Mohammad Amin Zaker
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran 11155-4563, Iran
| |
Collapse
|
10
|
Georgiou N, Kakava MG, Routsi EA, Petsas E, Stavridis N, Freris C, Zoupanou N, Moschovou K, Kiriakidi S, Mavromoustakos T. Quercetin: A Potential Polydynamic Drug. Molecules 2023; 28:8141. [PMID: 38138630 PMCID: PMC10745404 DOI: 10.3390/molecules28248141] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
The study of natural products as potential drug leads has gained tremendous research interest. Quercetin is one of those natural products. It belongs to the family of flavonoids and, more specifically, flavonols. This review summarizes the beneficial pharmaceutical effects of quercetin, such as its anti-cancer, anti-inflammatory, and antimicrobial properties, which are some of the quercetin effects described in this review. Nevertheless, quercetin shows poor bioavailability and low solubility. For this reason, its encapsulation in macromolecules increases its bioavailability and therefore pharmaceutical efficiency. In this review, a brief description of the different forms of encapsulation of quercetin are described, and new ones are proposed. The beneficial effects of applying new pharmaceutical forms of nanotechnology are outlined.
Collapse
Affiliation(s)
- Nikitas Georgiou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece; (N.G.); (E.A.R.); (E.P.); (N.S.); (N.Z.); (K.M.); (S.K.)
| | - Margarita Georgia Kakava
- Laboratory of Organic Chemistry and Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece;
| | - Efthymios Alexandros Routsi
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece; (N.G.); (E.A.R.); (E.P.); (N.S.); (N.Z.); (K.M.); (S.K.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Errikos Petsas
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece; (N.G.); (E.A.R.); (E.P.); (N.S.); (N.Z.); (K.M.); (S.K.)
| | - Nikolaos Stavridis
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece; (N.G.); (E.A.R.); (E.P.); (N.S.); (N.Z.); (K.M.); (S.K.)
| | - Christoforos Freris
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece;
| | - Nikoletta Zoupanou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece; (N.G.); (E.A.R.); (E.P.); (N.S.); (N.Z.); (K.M.); (S.K.)
| | - Kalliopi Moschovou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece; (N.G.); (E.A.R.); (E.P.); (N.S.); (N.Z.); (K.M.); (S.K.)
| | - Sofia Kiriakidi
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece; (N.G.); (E.A.R.); (E.P.); (N.S.); (N.Z.); (K.M.); (S.K.)
- Departamento de Quimica Orgánica, Facultade de Quimica, Universidade de Vigo, 36310 Vigo, Spain
| | - Thomas Mavromoustakos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece; (N.G.); (E.A.R.); (E.P.); (N.S.); (N.Z.); (K.M.); (S.K.)
| |
Collapse
|
11
|
Wang L, Chen M, Ran X, Tang H, Cao D. Sorafenib-Based Drug Delivery Systems: Applications and Perspectives. Polymers (Basel) 2023; 15:2638. [PMID: 37376284 DOI: 10.3390/polym15122638] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
As a Food and Drug Administration (FDA)-approved molecular-targeted chemotherapeutic drug, sorafenib (SF) can inhibit angiogenesis and tumor cell proliferation, leading to improved patient overall survival of hepatocellular carcinoma (HCC). In addition, SF is an oral multikinase inhibitor as a single-agent therapy in renal cell carcinoma. However, the poor aqueous solubility, low bioavailability, unfavorable pharmacokinetic properties and undesirable side effects (anorexia, gastrointestinal bleeding, and severe skin toxicity, etc.) seriously limit its clinical application. To overcome these drawbacks, the entrapment of SF into nanocarriers by nanoformulations is an effective strategy, which delivers SF in a target tumor with decreased adverse effects and improved treatment efficacy. In this review, significant advances and design strategies of SF nanodelivery systems from 2012 to 2023 are summarized. The review is organized by type of carriers including natural biomacromolecule (lipid, chitosan, cyclodextrin, etc.); synthetic polymer (poly(lactic-co-glycolic acid), polyethyleneimine, brush copolymer, etc.); mesoporous silica; gold nanoparticles; and others. Co-delivery of SF and other active agents (glypican-3, hyaluronic acid, apolipoprotein peptide, folate, and superparamagnetic iron oxide nanoparticles) for targeted SF nanosystems and synergistic drug combinations are also highlighted. All these studies showed promising results for targeted treatment of HCC and other cancers by SF-based nanomedicines. The outlook, challenges and future opportunities for the development of SF-based drug delivery are presented.
Collapse
Affiliation(s)
- Lingyun Wang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510641, China
| | - Meihuan Chen
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510641, China
| | - Xueguang Ran
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510641, China
| | - Hao Tang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510641, China
| | - Derong Cao
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510641, China
| |
Collapse
|
12
|
Pourmadadi M, Mahdi Eshaghi M, Ostovar S, Mohammadi Z, K. Sharma R, Paiva-Santos AC, Rahmani E, Rahdar A, Pandey S. Innovative nanomaterials for cancer diagnosis, imaging, and therapy: Drug deliveryapplications. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
|
13
|
Najafabadi AP, Pourmadadi M, Yazdian F, Rashedi H, Rahdar A, Díez-Pascual AM. pH-sensitive ameliorated quercetin delivery using graphene oxide nanocarriers coated with potential anticancer gelatin-polyvinylpyrrolidone nanoemulsion with bitter almond oil. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
14
|
Mahdi Eshaghi M, Pourmadadi M, Rahdar A, Díez-Pascual AM. Novel Carboxymethyl Cellulose-Based Hydrogel with Core-Shell Fe 3O 4@SiO 2 Nanoparticles for Quercetin Delivery. MATERIALS (BASEL, SWITZERLAND) 2022; 15:ma15248711. [PMID: 36556516 PMCID: PMC9784486 DOI: 10.3390/ma15248711] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/25/2022] [Accepted: 12/06/2022] [Indexed: 05/23/2023]
Abstract
A nanocomposite composed of carboxymethyl cellulose (CMC) and core-shell nanoparticles of Fe3O4@SiO2 was prepared as a pH-responsive nanocarrier for quercetin (QC) delivery. The nanoparticles were further entrapped in a water-in-oil-in-water emulsion system for a sustained release profile. The CMC/Fe3O4@SiO2/QC nanoparticles were characterized using dynamic light scattering (DLS), Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), a field emission scanning electron microscope (FE-SEM), and a vibrating sample magnetometer (VSM) to obtain insights into their size, stability, functional groups/chemical bonds, crystalline structure, morphology, and magnetic properties, respectively. The entrapment and loading efficiency were slightly improved after the incorporation of Fe3O4@SiO2 NPs within the hydrogel network. The dialysis method was applied for drug release studies. It was found that the amount of QC released increased with the decrease in pH from 7.4 to 5.4, while the sustained-release pattern was preserved. The A549 cell line was chosen to assess the anticancer activity of the CMC/Fe3O4@SiO2/QC nanoemulsion and its components for lung cancer treatment via an MTT assay. The L929 cell line was used in the MTT assay to determine the possible side effects of the nanoemulsion. Moreover, a flow cytometry test was performed to measure the level of apoptosis and necrosis. Based on the obtained results, CMC/Fe3O4@SiO2 can be regarded as a novel promising system for cancer therapy.
Collapse
Affiliation(s)
- Mohammad Mahdi Eshaghi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Mehrab Pourmadadi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 1417935840, Iran
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol 538-98615, Iran
| | - Ana M. Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain
| |
Collapse
|