1
|
Rehan F, Karim ME, Ahemad N, Qureshi OS, Jelani S, Gan SH, Chowdhury EH. Modified sodium caseinate-based nanomicelles for enhanced chemotherapeutics against breast cancer via improved cellular uptake and cytotoxicity. Drug Dev Ind Pharm 2025:1-18. [PMID: 40265846 DOI: 10.1080/03639045.2025.2495849] [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: 09/02/2024] [Revised: 03/05/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE Poor prognosis, drug resistance, and lower drug loading capacity of the delivery systems lead to therapeutic failures of breast cancers. Herein, we functionalized sodium caseinate nanomicelles (NaCNs) with the divalent calcium (Ca2+) and the glucose (Glc) to increase the loading capacity of micelles for higher cellular uptake and cytotoxicity against breast cancer cells. METHODOLOGY Modification of casein micelles was confirmed through Fourier transform infrared spectra (FTIR). Triple quadrupole liquid chromatography-mass spectrometry (TQOF-LCMS/MS) was utilized as a simple, rapid, and sensitive method for protein corona quantification around casein through SwissProt.Mus_musculus database and through de novo sequencing. Un-modified and modified casein micelles were further characterized through field emission scanning electron microscope (FESEM), high resolution-transmission electron microscope (HR-TEM), and energy-dispersive X-ray (EDX). Whereas, sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) was used for protein separation and analysis during micelles formation. RESULTS Calcium divalent modified sodium caseinate nanomicelles (Ca-NaCNs) and glucose-modified sodium caseinate nanomicelles (Glc-NaCNs) were successfully developed, demonstrating a significantly improved micellar stability. Glc-NaCNs-DOX showed a zeta size of 297.13 ± 15.66 nm with an improved zeta potential of -13.73 ± 0.579 with a drug loading efficiency (DLE) of 86% as compared to our previously published casein formulations since the modified versions involved more soluble casein in the protein micelle matrix, Whereas, Ca-NaCNs-DOX also showed an IC50 value of approximately 197.1 nm as compared to IC50 of free DOX (341.8 nm) and when compared to unmodified DOX loaded formulations (p < .001). CONCLUSION Modified NaCNs exhibit the potential to be investigated further as a novel delivery system for similar active moieties to maximize their therapeutic effects.
Collapse
Affiliation(s)
- Farah Rehan
- School of Pharmacy, Monash University Malaysia, Petaling Jaya, Malaysia
- Department of Molecular Medicine and Al-Jawhara Centre for Molecular Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Md Emranul Karim
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Petaling Jaya, Malaysia
| | - Omer Salman Qureshi
- Department of Pharmacy, Forman Christian College University, Lahore, Pakistan
| | - Seemal Jelani
- Department of Chemistry, Forman Christian College University, Lahore, Pakistan
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Petaling Jaya, Malaysia
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Selangor Darul Ehsan, Malaysia
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya, Malaysia
- Daffodil International University, Birulia, Bangladesh
- Nanoflex LLC, Leesburg, FL, USA
| |
Collapse
|
2
|
Kleszcz R, Majchrzak-Celińska A, Baer-Dubowska W. Tannins in cancer prevention and therapy. Br J Pharmacol 2025; 182:2075-2093. [PMID: 37614022 DOI: 10.1111/bph.16224] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/31/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Tannins are a heterogenous class of polyphenolic natural products with promising cancer chemopreventive and therapeutic potential. Studies undertaken over the last 30 years have demonstrated their capacity to target many cellular pathways and molecules important in the development of cancer. Recently, new mechanisms that might be important in anti-carcinogenic activity, such as inhibition of epithelial-to-mesenchymal transition, reduction of cancer stem cell creation, and modulation of cancer cells metabolism have been described. Along with the mechanisms underlying the anti-cancer activity of tannins, this review focuses on their possible application as chemosensitizers in adjuvant therapy and countering multidrug resistance. Furthermore, characteristic physicochemical properties of some tannins, particularly tannic acid, are useful in the formation of nanovehicles for anticancer drugs or the isolation of circulating cancer cells. These new potential applications of tannins deserve further studies. Well-designed clinical trials, which are scarce, are needed to assess the therapeutic effects of tannins themselves or as adjuvants in cancer treatment. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Robert Kleszcz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznań, Poland
| | | | - Wanda Baer-Dubowska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
3
|
Moutaoukil ME, Lolli MG, D'Amone S, Khan M, Grillo R, Eyer J, Grieco M, Ursini O, Spadavecchia J, Cortese B. Doxorubicin and NFL-TBS.40-63 peptide loaded gold nanoparticles as a multimodal therapy of glioblastoma. DISCOVER NANO 2025; 20:72. [PMID: 40293574 PMCID: PMC12037963 DOI: 10.1186/s11671-025-04249-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025]
Abstract
Conventional treatments for glioblastoma (GBM) are hindered by systemic toxicity, limited blood-brain barrier penetration, and therapeutic resistance. To address these challenges, we developed dual-functionalized gold nanoparticles (AuNPs) conjugated with a biotinylated NFL-TBS.40-63 peptide and the chemotherapeutic agent doxorubicin. This platform integrates targeted delivery and therapeutic action to enhance efficacy while minimising off-target effects. Our findings reveal superior cellular uptake, dose- and time-dependent cytotoxicity, and apoptosis induction in GBM cells compared to mono-functionalized counterparts. Furthermore, pH-sensitive drug release profiles underscore the system's potential to exploit the tumour microenvironment's acidic conditions for precise drug delivery. Comprehensive characterisation confirmed the stability, biocompatibility, and functional efficacy of the dual-functionalized AuNPs. This study highlights the promise of these nanoconjugates as a multimodal approach to GBM therapy, paving the way for further translational research in nanomedicine.
Collapse
Affiliation(s)
- Myriam El Moutaoukil
- CNRS, NBD-CSPBAT, Laboratory of Chemistry, Structures and Properties of Biomaterials and Therapeutic Agents University Paris13, Sorbonne Paris Nord, Bobigny, France
| | - Maria Grazia Lolli
- National Research Council - Institute of Nanotechnology (CNR Nanotec), c/o Department of Physics "E. Fermi", University Sapienza, Pz.le Aldo Moro 5, 00185, Rome, Italy
| | - Stefania D'Amone
- National Research Council - Institute of Nanotechnology (CNR Nanotec), c/o Ecotekne, University of Salento, Via Monteroni, 73100, Lecce, Italy
| | - Memona Khan
- CNRS, NBD-CSPBAT, Laboratory of Chemistry, Structures and Properties of Biomaterials and Therapeutic Agents University Paris13, Sorbonne Paris Nord, Bobigny, France
| | - Roberta Grillo
- National Research Council - Institute of Nanotechnology (CNR Nanotec), c/o Department of Physics "E. Fermi", University Sapienza, Pz.le Aldo Moro 5, 00185, Rome, Italy
| | - Joel Eyer
- Laboratoire Micro et NanomedecinesTranslationnelles, Inserm 1066, CNRS 6021, Institut de Recherche enIngénierie de la Sante, Bâtiment IBS Institut de Biologie de La Sante, Université´ Angers, Centre Hospitalier Universitaire, 49100, Angers, France
| | - Maddalena Grieco
- National Research Council - Institute of Nanotechnology (CNR Nanotec), c/o Ecotekne, University of Salento, Via Monteroni, 73100, Lecce, Italy
| | - Ornella Ursini
- National Research Council - Institute of Nanotechnology (CNR Nanotec), c/o Department of Physics "E. Fermi", University Sapienza, Pz.le Aldo Moro 5, 00185, Rome, Italy
| | - Jolanda Spadavecchia
- CNRS, UMR 7244, NBD-CSPBAT, Laboratory of Chemistry, Structures and Properties of Biomaterials and Therapeutic Agents University Paris13, Sorbonne Paris Nord, Bobigny, France
| | - Barbara Cortese
- National Research Council - Institute of Nanotechnology (CNR Nanotec), c/o Department of Physics "E. Fermi", University Sapienza, Pz.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
4
|
Akdaşçi E, Eker F, Duman H, Singh P, Bechelany M, Karav S. Lactoferrin as a Versatile Agent in Nanoparticle Applications: From Therapeutics to Agriculture. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:2018. [PMID: 39728554 PMCID: PMC11728633 DOI: 10.3390/nano14242018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
Nanoparticles (NPs) have emerged as a potent choice for various applications, from drug delivery to agricultural studies, serving as an alternative and promising methodology for future advancements. They have been widely explored in delivery systems, demonstrating immense promise and high efficiency for the delivery of numerous biomolecules such as proteins and anticancer agents, either solely or modified with other compounds to enhance their capabilities. In addition, the utilization of NPs extends to antimicrobial studies, where they are used to develop novel antibacterial, antifungal, and antiviral formulations with advanced characteristics. Lactoferrin (Lf) is a glycoprotein recognized for its significant multifunctional properties, such as antimicrobial, antioxidant, anti-inflammatory, anticancer, and neuroprotective effects. Its activity has a broad distribution in the human body, with Lf receptors present in multiple regions. Current research shows that Lf is utilized in NP technology as a surface material, encapsulated biomolecule, and even as an NP itself. Due to the abundance of Lf receptors in various regions, Lf can be employed as a surface material in NPs for targeted delivery strategies, particularly in crossing the BBB and targeting specific cancers. Furthermore, Lf can be synthesized in an NP structure, positioning it as a strong candidate in future NP-related applications. In this article, we explore the highlighted and underexplored areas of Lf applications in NPs research.
Collapse
Affiliation(s)
- Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye; (E.A.); (F.E.); (H.D.)
| | - Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye; (E.A.); (F.E.); (H.D.)
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye; (E.A.); (F.E.); (H.D.)
| | - Priyanka Singh
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kongens Lyngby, Denmark;
| | - Mikhael Bechelany
- Institut Européen des Membranes (IEM), UMR 5635, University Montpellier, École Nationale Supérieure de Chimie de Montpellier (ENSCM), Centre National de la Recherche Scientifique (CNRS), F-34095 Montpellier, France
- Functional Materials Group, Gulf University for Science and Technology (GUST), Masjid Al Aqsa Street, Mubarak Al-Abdullah 32093, Kuwait
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye; (E.A.); (F.E.); (H.D.)
| |
Collapse
|
5
|
Zhang D, Yuan Y, Xiong J, Zeng Q, Gan Y, Jiang K, Xie N. Anti-breast cancer effects of dairy protein active peptides, dairy products, and dairy protein-based nanoparticles. Front Pharmacol 2024; 15:1486264. [PMID: 39605907 PMCID: PMC11598434 DOI: 10.3389/fphar.2024.1486264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Breast cancer is the most frequently diagnosed and fatal cancer among women worldwide. Dairy protein-derived peptides and dairy products are important parts of the daily human diet and have shown promising activities in suppressing the proliferation, migration, and invasion of breast cancer cells, both in vitro and in vivo. Most of the review literature employs meta-analysis methods to explore the association between dairy intake and breast cancer risk. However, there is a lack of comprehensive summary regarding the anti-breast cancer properties of dairy protein-derived peptides, dairy products, and dairy protein-based nanoparticles as well as their underlying mechanisms of action. Therefore, the present study discussed the breast cancer inhibitory effects and mechanisms of active peptides derived from various dairy protein sources. Additionally, the characteristics, anti-breast cancer activities and active components of several types of dairy products, including fermented milk, yogurt and cheeses, were summarized. Furthermore, the preparation methods and therapeutic effects of various dairy protein-containing nanoparticle delivery systems for breast cancer therapy were briefly described. Lastly, this work also provided an overview of what is currently known about the anti-breast cancer effects of dairy products in clinical studies. Our review will be of interest to the development of natural anticancer drugs.
Collapse
Affiliation(s)
- Deju Zhang
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Ying Yuan
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Juan Xiong
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qingdong Zeng
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yiming Gan
- Plant Science, School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kai Jiang
- Eastern Institute for Advanced Study, Eastern Institute of Technology, Ningbo, Zhejiang, China
| | - Ni Xie
- Biobank, Shenzhen Second People’s Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
6
|
Nashaat Alnagar A, Motawea A, Elamin KM, Abu Hashim II. Hyaluronic acid/lactoferrin-coated polydatin/PLGA nanoparticles for active targeting of CD44 receptors in lung cancer. Pharm Dev Technol 2024; 29:1016-1032. [PMID: 39392049 DOI: 10.1080/10837450.2024.2414937] [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: 05/25/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Traditional chemotherapeutic drugs lack optimal efficacy and invoke severe adverse effects in cancer patients. Polydatin (PD), a phytomedicine, has gradually gained attention due to its antitumor activity. However, its low solubility and poor bioavailability are still cornerstone issues. The present study aimed to fabricate and develop hyaluronic acid/lactoferrin-double coated PD/PLGA nanoparticles via a layer-by-layer self-assembly technique for active targeting of CD44 receptors in lung cancer. Different molecular weights (M.wt.) of HA (32 and 110 kDa) were exploited to study the relationship between the HA M.wt. and the NPs targeting efficacy. The optimized formulations were fully characterized. Their cytotoxicity and cellular uptake were investigated against A549 cell line by CCK-8 kit and fluorescence imaging, respectively. Finally, HA110/Lf-coated PD/PLGA NPs (F9) were subjected to a competitive inhibition study to prove internalization through CD44 overexpressed receptors. The results verified the fabrication of F9 with a particle size of 174.87 ± 3.97 nm and a zeta potential of -24.37 ± 1.19 mV as well as spherical NPs architecture. Importantly, it provoked enhanced cytotoxicity (IC50 = 0.57 ± 0.02 µg/mL) and superior cellular uptake efficacy. To conclude, the current investigation lays the foundation for the prospective therapeutic avenue of F9 for active targeting of CD44 receptors in lung cancer.
Collapse
Affiliation(s)
- Ahmed Nashaat Alnagar
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia, Egypt
| | - Amira Motawea
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia, Egypt
| | - Khaled M Elamin
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Irhan Ibrahim Abu Hashim
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia, Egypt
| |
Collapse
|
7
|
Marwaha D, Singh N, Gautam S, Rai N, Tiwari P, Bakshi AK, Kumar A, Agarwal N, Sharma M, Shukla RP, Mishra PR. Pemetrexed-loaded supramolecular acetal-functionalized pH-responsive nanocarriers selectively induce apoptosis through biotin receptors to enhance antitumor efficacy. Colloids Surf B Biointerfaces 2024; 245:114247. [PMID: 39368420 DOI: 10.1016/j.colsurfb.2024.114247] [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: 03/01/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024]
Abstract
A novel pH-responsive crystalsomes has been developed using acetal-functionalized pillar[5]arenes (AP[5]) and methyl viologen (MV) through host-guest interactions. The successful synthesis of AP[5] was confirmed via 1H-NMR spectroscopy, while the formation of the host-guest complex between AP[5] and MV was also verified using ¹H-NMR. The supramolecular assemblies formed at a 1:1 molar ratio of AP[5] to MV exhibited remarkable colloidal stability, a negative surface charge, and a high association constant.An acetal-functionalized pillara[5]arenes (AP[5]) crystalsomes were fabricated to reduce the toxicity of pemetrexed (PMX) in off-target sites and deliver the therapeutic doses to the active sites. Extensive characterization of the crystalsomes was performed, revealing their morphology and crystalline structure through SEM and TEM imaging. WAXS analysis confirmed the crystalline nature of the assemblies, and SAED patterns indicated that the crystalsome shell consisted of lamellae resembling single crystals with polymer chains oriented parallel to the interface. To enhnace the targeting capabilities, the surface of the crystalsomes was modified with biotin by conjugating viologen with biotin (MV-BT), aiming to target biotin receptors overexpressed on tumor cells. These biotin -modified crystalsomes (PMX-BT@CLs) were designed to be acid-labile facilitating the release of encapsulated drugs upon cellular internalization, as confirmed by confocal laser scanning microscopy (CLSM). In vivo, studies demonstrated that the PMX-loaded crystalsomes remained in circulation for extended period, showing improved pharmacokinetics. The area under the curve (AUC) of PMX-BT@CLs was approxiately 3.9 times higher than that of the free drug. Additionally, the relative tumor volume was found to be about 3.5 times lower in the group treated with biotin-modified crystalsomes compared to those treated with free PMX. The mean survival time was also significantly enhanced in the PMX-BT@CLs group. This study underscores the potential of using host-guest motifs in drug delivery app;ications, demonstrating the PMX can effectively targted to tumor sites with minimal off-target toxicity.
Collapse
Affiliation(s)
- Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, UP 201002, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Ankit Kumar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, UP 201002, India.
| |
Collapse
|
8
|
Pradeep Prabhu P, Mohanty B, Lobo CL, Balusamy SR, Shetty A, Perumalsamy H, Mahadev M, Mijakovic I, Dubey A, Singh P. Harnessing the nutriceutics in early-stage breast cancer: mechanisms, combinational therapy, and drug delivery. J Nanobiotechnology 2024; 22:574. [PMID: 39294665 PMCID: PMC11411841 DOI: 10.1186/s12951-024-02815-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is a significant health challenge, ranking as the second leading cause of cancer-related death and the primary cause of mortality among women aged 45 to 55. Early detection is crucial for optimal prognosis. Among various treatment options available for cancer, chemotherapy remains the predominant approach. However, its patient-friendliness is hindered by cytotoxicity, adverse effects, multi-drug resistance, potential for recurrence, and high costs. This review explores extensively studied phytomolecules, elucidating their molecular mechanisms. It also emphasizes the importance of combination therapy, highlighting recent advancements in the exploration of diverse drug delivery systems and novel routes of administration. The regulatory considerations are crucial in translating these approaches into clinical practices. RESULTS Consequently, there is growing interest in exploring the relationship between diet, cancer, and complementary and alternative medicine (CAM) in cancer chemotherapy. Phytochemicals like berberine, curcumin, quercetin, lycopene, sulforaphane, resveratrol, epigallocatechin gallate, apigenin, genistein, thymoquinone have emerged as promising candidates due to their pleiotropic actions on target cells through multiple mechanisms with minimal toxicity effects. This review focuses on extensively studied phytomolecules, elucidating their molecular mechanisms. It also emphasizes the importance of combination therapy, highlighting recent advancements in the exploration of diverse drug delivery systems and novel routes of administration. The regulatory considerations are crucial in translating these approaches into clinical practices. CONCLUSION The present review provides a comprehensive understanding of the molecular mechanisms, coupled with well-designed clinical trials and adherence to regulatory guidelines, which pave the way for nutrition-based combination therapies to become a frontline approach in early-stage BC treatment.
Collapse
Affiliation(s)
- Pavithra Pradeep Prabhu
- Nitte (Deemed to Be University), Department of Pharmacognosy, NGSM Institute of Pharmaceutical Sciences, Mangaluru, 575018, India
| | - Barsha Mohanty
- Nitte (Deemed to Be University), Department of Molecular Genetics and Cancer Biology, Nitte University Centre for Science, Education and Research, Mangaluru, 575018, India
| | - Cynthia Lizzie Lobo
- Nitte (Deemed to Be University), Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Mangaluru, 575018, India
| | - Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University, Gwangjin-Gu, Seoul, Republic of Korea.
| | - Amitha Shetty
- Nitte (Deemed to Be University), Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Mangaluru, 575018, India
| | - Haribalan Perumalsamy
- Center for Creative Convergence Education, Hanyang University, Seoul, Republic of Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, South Korea, Hanyang University, Seoul, Republic of Korea
| | - Manohar Mahadev
- Nitte (Deemed to Be University), Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Mangaluru, 575018, India
| | - Ivan Mijakovic
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Akhilesh Dubey
- Nitte (Deemed to Be University), Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Mangaluru, 575018, India.
| | - Priyanka Singh
- Systems and Synthetic Biology Division, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden.
| |
Collapse
|
9
|
Li H, Guan M, Zhang NN, Wang Y, Liang T, Wu H, Wang C, Sun T, Liu S. Harnessing nanomedicine for modulating microglial states in the central nervous system disorders: Challenges and opportunities. Biomed Pharmacother 2024; 177:117011. [PMID: 38917758 DOI: 10.1016/j.biopha.2024.117011] [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: 03/12/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Microglia are essential for maintaining homeostasis and responding to pathological events in the central nervous system (CNS). Their dynamic and multidimensional states in different environments are pivotal factors in various CNS disorders. However, therapeutic modulation of microglial states is challenging due to the intricate balance these cells maintain in the CNS environment and the blood-brain barrier's restriction of drug delivery. Nanomedicine presents a promising avenue for addressing these challenges, offering a method for the targeted and efficient modulation of microglial states. This review covers the challenges faced in microglial therapeutic modulation and potential use of nanoparticle-based drug delivery systems. We provide an in-depth examination of nanoparticle applications for modulating microglial states in a range of CNS disorders, encompassing neurodegenerative and autoimmune diseases, infections, traumatic injuries, stroke, tumors, chronic pain, and psychiatric conditions. This review highlights the recent advancements and future prospects in nanomedicine for microglial modulation, paving the way for future research and clinical applications of therapeutic interventions in CNS disorders.
Collapse
Affiliation(s)
- Haisong Li
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Meng Guan
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ning-Ning Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China
| | - Yizhuo Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tingting Liang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Haitao Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chang Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Shuhan Liu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| |
Collapse
|
10
|
Hashemi M, Khosroshahi EM, Chegini MK, Asadi S, Hamyani Z, Jafari YA, Rezaei F, Eskadehi RK, Kojoori KK, Jamshidian F, Nabavi N, Alimohammadi M, Rashidi M, Mahmoodieh B, Khorrami R, Taheriazam A, Entezari M. Mechanistic insights into cisplatin response in breast tumors: Molecular determinants and drug/nanotechnology-based therapeutic opportunities. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108513. [PMID: 39216513 DOI: 10.1016/j.mrrev.2024.108513] [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: 05/12/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer continues to be a major global health challenge, driving the need for effective therapeutic strategies. Cisplatin, a powerful chemotherapeutic agent, is widely used in breast cancer treatment. However, its effectiveness is often limited by systemic toxicity and the development of drug resistance. This review examines the molecular factors that influence cisplatin response and resistance, offering crucial insights for the scientific community. It highlights the significance of understanding cisplatin resistance's genetic and epigenetic contributors, which could lead to more personalized treatment approaches. Additionally, the review explores innovative strategies to counteract cisplatin resistance, including combination therapies, nanoparticle-based drug delivery systems, and targeted therapies. These approaches are under intensive investigation and promise to enhance breast cancer treatment outcomes. This comprehensive discussion is a valuable resource to advance breast cancer therapeutics and address the challenge of cisplatin resistance.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrnaz Kalhor Chegini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Hamyani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Medicine, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran
| | - Yasamin Alsadat Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rezaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ramtin Khodaparast Eskadehi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kimia Kia Kojoori
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Faranak Jamshidian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Rashidi
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
11
|
Rahimkhoei V, Alzaidy AH, Abed MJ, Rashki S, Salavati-Niasari M. Advances in inorganic nanoparticles-based drug delivery in targeted breast cancer theranostics. Adv Colloid Interface Sci 2024; 329:103204. [PMID: 38797070 DOI: 10.1016/j.cis.2024.103204] [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: 10/12/2023] [Revised: 04/10/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Theranostic nanoparticles (NPs) have the potential to dramatically improve cancer management by providing personalized medicine. Inorganic NPs have attracted widespread interest from academic and industrial communities because of their unique physicochemical properties (including magnetic, thermal, and catalytic performance) and excellent functions with functional surface modifications or component dopants (e.g., imaging and controlled release of drugs). To date, only a restricted number of inorganic NPs are deciphered into clinical practice. This review highlights the recent advances of inorganic NPs in breast cancer therapy. We believe that this review can provides various approaches for investigating and developing inorganic NPs as promising compounds in the future prospects of applications in breast cancer treatment and material science.
Collapse
Affiliation(s)
- Vahid Rahimkhoei
- Institute of Nano Science and Nano Technology, University of Kashan, P.O. Box 87317-51167, Kashan, Islamic Republic of Iran
| | - Asaad H Alzaidy
- Department of Laboratory and Clinical Science, College of Pharmacy, University of Al-Qadisiyah, Diwaniyah, Iraq
| | - May Jaleel Abed
- Department of Chemistry, College of Education, University of Al-Qadisiyah, Diwaniyah, Iraq
| | - Somaye Rashki
- Department of Microbiology, Iranshahr University of Medical Sciences, Iranshahr, Islamic Republic of Iran
| | - Masoud Salavati-Niasari
- Institute of Nano Science and Nano Technology, University of Kashan, P.O. Box 87317-51167, Kashan, Islamic Republic of Iran.
| |
Collapse
|
12
|
Attri K, Chudasama B, Mahajan RL, Choudhury D. Integrated insulin-iron nanoparticles: a multi-modal approach for receptor-specific bioimaging, reactive oxygen species scavenging, and wound healing. DISCOVER NANO 2024; 19:96. [PMID: 38814485 PMCID: PMC11139842 DOI: 10.1186/s11671-024-04024-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/26/2024] [Indexed: 05/31/2024]
Abstract
Metallic nanoparticles have emerged as a promising option for various biological applications, owing to their distinct characteristics such as small size, optical properties, and ability to exhibit luminescence. In this study, we have successfully employed a one-pot method to synthesize multifunctional insulin-protected iron [Fe(II)] nanoparticles denoted as [IFe(II)NPs]. The formation of IFe(II)NPs is confirmed by the presence of FTIR bonds at 447.47 and 798.28 cm-1, corresponding to Fe-O and Fe-N bonds, respectively. Detailed analysis of the HR-TEM-EDS-SAED data reveals that the particles are spherical in shape, partially amorphous in nature, and have a diameter of 28.6 ± 5.2 nm. Additionally, Metal Ion Binding (MIB) and Protein Data Bank (PDB) analyses affirm the binding of iron ions to the insulin hexamer. Our findings underscore the potential of IFe(II)NPs as a promising new platform for a variety of biomedical applications due to their high signal-to-noise ratio, and minimal background fluorescence. The particles are highly luminescent, biocompatible, and have a significant quantum yield (0.632). Exemplar applications covered in this paper include insulin receptor recognition and protection against reactive oxygen species (ROS), harmful molecules known to inflict damage on cells and DNA. The IFe(II)NPs effectively mitigate ROS-induced inflammation, which is a hinderance to wound recovery, thereby facilitating enhanced wound recovery.
Collapse
Affiliation(s)
- Komal Attri
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
- Centre of Excellence for Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Bhupendra Chudasama
- Centre of Excellence for Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Department of Physics and Material Sciences, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| | - Roop L Mahajan
- Department of Physics and Material Sciences, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Department of Mechanical Engineering, Department of Materials Science and Engineering Virginia Tech, Blacksburg, VA, 24061, USA.
| | - Diptiman Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Centre of Excellence for Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
13
|
Manzari‐Tavakoli A, Babajani A, Tavakoli MM, Safaeinejad F, Jafari A. Integrating natural compounds and nanoparticle-based drug delivery systems: A novel strategy for enhanced efficacy and selectivity in cancer therapy. Cancer Med 2024; 13:e7010. [PMID: 38491817 PMCID: PMC10943377 DOI: 10.1002/cam4.7010] [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: 10/24/2023] [Revised: 12/05/2023] [Accepted: 12/10/2023] [Indexed: 03/18/2024] Open
Abstract
Cancer remains a leading cause of death worldwide, necessitating the development of innovative and more effective treatment strategies. Conventional cancer treatments often suffer from limitations such as systemic toxicity, poor pharmacokinetics, and drug resistance. Recently, there has been growing attention to utilizing natural compounds derived from various sources as possible cancer therapeutics. Natural compounds have demonstrated diverse bioactive properties, including antioxidant, anti-inflammatory, and antitumor effects, making them attractive candidates for cancer treatment. However, their limited solubility and bioavailability present challenges for effective delivery to cancer cells. To overcome these limitations, researchers have turned to nanotechnology-based drug delivery systems. Nanoparticles, with their small size and unique properties, can encapsulate therapeutic agents and offer benefits such as improved solubility, prolonged drug release, enhanced cellular uptake, and targeted delivery. Functionalizing nanoparticles with specific ligands further enhances their precision in recognizing and binding to cancer cells. Combining natural compounds with nanotechnology holds great promise in achieving efficient and safe cancer treatments by enhancing bioavailability, pharmacokinetics, and selectivity toward cancer cells. This review article provides an overview of the advancements in utilizing natural substances and nanotechnology-based drug delivery systems for cancer treatment. It discusses the benefits and drawbacks of various types of nanoparticles, as well as the characteristics of natural compounds that make them appealing for cancer therapy. Additionally, current research on natural substances and nanoparticles in preclinical and clinical settings is highlighted. Finally, the challenges and future perspectives in developing natural compound-nanoparticle-based cancer therapies are discussed.
Collapse
Affiliation(s)
| | - Amirhesam Babajani
- Oncopathology Research Center, Department of Molecular Medicine, School of MedicineIran University of Medical SciencesTehranIran
| | - Maryam Manzari Tavakoli
- Department of PhytochemistryMedicinal Plants and Drugs Research Institute, Shahid Beheshti UniversityTehranIran
| | - Fahimeh Safaeinejad
- Traditional Medicine and Materia Medica Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Ameneh Jafari
- Chronic Respiratory Diseases Research Center, NRITLDShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
14
|
Golmohammadi M, Zamanian MY, Jalal SM, Noraldeen SAM, Ramírez‐Coronel AA, Oudaha KH, Obaid RF, Almulla AF, Bazmandegan G, Kamiab Z. A comprehensive review on Ellagic acid in breast cancer treatment: From cellular effects to molecular mechanisms of action. Food Sci Nutr 2023; 11:7458-7468. [PMID: 38107139 PMCID: PMC10724635 DOI: 10.1002/fsn3.3699] [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: 05/17/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 10/16/2023] Open
Abstract
Globally, breast cancer (BC) is the leading cause of cancer-related deaths in women. Hence, developing a therapeutic plan to overcome the disease is crucial. Numerous factors such as endogenous hormones and environmental factors may play a role in the pathophysiology of BC. Regarding the multi-modality treatment of BC, natural compounds like ellagic acid (EA) received has received increased interest in antitumor efficacy with lower adverse effects. Based on the results of this comprehensive review, EA has multiple effects on BC cells including (1) suppresses the growth of BC cells by arresting the cell cycle in the G0/G1 phase, (2) suppresses migration, invasion, and metastatic, (3) stimulates apoptosis in MCF-7 cells via TGF-β/Smad3 signaling axis, (4) inhibits CDK6 that is important in cell cycle regulation, (5) binds to ACTN4 and induces its degradation via the ubiquitin-proteasome pathway, inducing decreased cell motility and invasion in BC cells, (6) inhibits the PI3K/AKT pathway, and (7) inhibits angiogenesis-associated activities including proliferation (reduces VEGFR-2 tyrosine kinase activity). In conclusion, EA exhibits anticancer activity through various molecular mechanisms that influence key cellular processes like apoptosis, cell cycle, angiogenesis, and metastasis in BC. However, further researches are essential to fully elucidate its molecular targets and implications for clinical applications.
Collapse
Affiliation(s)
| | - Mohammad Yasin Zamanian
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and Toxicology, School of PharmacyHamadan University of Medical SciencesHamadanIran
| | | | | | - Andrés Alexis Ramírez‐Coronel
- Research Group in Educational StatisticsNational University of Education (UNAE)AzoguesEcuador
- Epidemiology and Biostatistics Research GroupCES UniversityMedellínColombia
| | - Khulood H. Oudaha
- Pharmaceutical Chemistry Department, College of PharmacyAl‐Ayen UniversityThi‐OarIraq
| | - Rasha Fadhel Obaid
- Department of Biomedical EngineeringAl‐Mustaqbal University CollegeBabylonIraq
| | - Abbas F. Almulla
- Department of Medical Laboratory Technology, College of Medical TechnologyIslamic UniversityNajafIraq
| | - Gholamreza Bazmandegan
- Physiology‐Pharmacology Research Center, Research Institute of Basic Medical SciencesRafsanjan University of Medical SciencesRafsanjanIran
- Department of Physiology and Pharmacology, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Zahra Kamiab
- Clinical Research Development Unit, Ali‐Ibn Abi‐Talib HospitalRafsanjan University of Medical SciencesRafsanjanIran
- Department of Community Medicine, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| |
Collapse
|
15
|
Kaymaz SV, Nobar HM, Sarıgül H, Soylukan C, Akyüz L, Yüce M. Nanomaterial surface modification toolkit: Principles, components, recipes, and applications. Adv Colloid Interface Sci 2023; 322:103035. [PMID: 37931382 DOI: 10.1016/j.cis.2023.103035] [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: 07/23/2023] [Revised: 09/11/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
Surface-functionalized nanostructures are at the forefront of biotechnology, providing new opportunities for biosensors, drug delivery, therapy, and bioimaging applications. The modification of nanostructures significantly impacts the performance and success of various applications by enabling selective and precise targeting. This review elucidates widely practiced surface modification strategies, including click chemistry, cross-coupling, silanization, aldehyde linkers, active ester chemistry, maleimide chemistry, epoxy linkers, and other protein and DNA-based methodologies. We also delve into the application-focused landscape of the nano-bio interface, emphasizing four key domains: therapeutics, biosensing, environmental monitoring, and point-of-care technologies, by highlighting prominent studies. The insights presented herein pave the way for further innovations at the intersection of nanotechnology and biotechnology, providing a useful handbook for beginners and professionals. The review draws on various sources, including the latest research articles (2018-2023), to provide a comprehensive overview of the field.
Collapse
Affiliation(s)
- Sümeyra Vural Kaymaz
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey; SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | | | - Hasan Sarıgül
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | - Caner Soylukan
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | - Lalehan Akyüz
- Department of Molecular Biology and Genetics, Aksaray University, 68100 Aksaray, Turkey
| | - Meral Yüce
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey.
| |
Collapse
|
16
|
Ajith S, Almomani F, Elhissi A, Husseini GA. Nanoparticle-based materials in anticancer drug delivery: Current and future prospects. Heliyon 2023; 9:e21227. [PMID: 37954330 PMCID: PMC10637937 DOI: 10.1016/j.heliyon.2023.e21227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 09/18/2023] [Accepted: 10/18/2023] [Indexed: 11/14/2023] Open
Abstract
The past decade has witnessed a breakthrough in novel strategies to treat cancer. One of the most common cancer treatment modalities is chemotherapy which involves administering anti-cancer drugs to the body. However, these drugs can lead to undesirable side effects on healthy cells. To overcome this challenge and improve cancer cell targeting, many novel nanocarriers have been developed to deliver drugs directly to the cancerous cells and minimize effects on the healthy tissues. The majority of the research studies conclude that using drugs encapsulated in nanocarriers is a much safer and more effective alternative than delivering the drug alone in its free form. This review provides a summary of the types of nanocarriers mainly studied for cancer drug delivery, namely: liposomes, polymeric micelles, dendrimers, magnetic nanoparticles, mesoporous nanoparticles, gold nanoparticles, carbon nanotubes and quantum dots. In this review, the synthesis, applications, advantages, disadvantages, and previous studies of these nanomaterials are discussed in detail. Furthermore, the future opportunities and possible challenges of translating these materials into clinical applications are also reported.
Collapse
Affiliation(s)
- Saniha Ajith
- Department of Chemical Engineering, College of Engineering, Qatar University, Doha, Qatar
| | - Fares Almomani
- Department of Chemical Engineering, College of Engineering, Qatar University, Doha, Qatar
| | | | - Ghaleb A. Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| |
Collapse
|
17
|
Ashrafizadeh M, Zarrabi A, Bigham A, Taheriazam A, Saghari Y, Mirzaei S, Hashemi M, Hushmandi K, Karimi-Maleh H, Nazarzadeh Zare E, Sharifi E, Ertas YN, Rabiee N, Sethi G, Shen M. (Nano)platforms in breast cancer therapy: Drug/gene delivery, advanced nanocarriers and immunotherapy. Med Res Rev 2023; 43:2115-2176. [PMID: 37165896 DOI: 10.1002/med.21971] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/09/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023]
Abstract
Breast cancer is the most malignant tumor in women, and there is no absolute cure for it. Although treatment modalities including surgery, chemotherapy, and radiotherapy are utilized for breast cancer, it is still a life-threatening disease for humans. Nanomedicine has provided a new opportunity in breast cancer treatment, which is the focus of the current study. The nanocarriers deliver chemotherapeutic agents and natural products, both of which increase cytotoxicity against breast tumor cells and prevent the development of drug resistance. The efficacy of gene therapy is boosted by nanoparticles and the delivery of CRISPR/Cas9, Noncoding RNAs, and RNAi, promoting their potential for gene expression regulation. The drug and gene codelivery by nanoparticles can exert a synergistic impact on breast tumors and enhance cellular uptake via endocytosis. Nanostructures are able to induce photothermal and photodynamic therapy for breast tumor ablation via cell death induction. The nanoparticles can provide tumor microenvironment remodeling and repolarization of macrophages for antitumor immunity. The stimuli-responsive nanocarriers, including pH-, redox-, and light-sensitive, can mediate targeted suppression of breast tumors. Besides, nanoparticles can provide a diagnosis of breast cancer and detect biomarkers. Various kinds of nanoparticles have been employed for breast cancer therapy, including carbon-, lipid-, polymeric- and metal-based nanostructures, which are different in terms of biocompatibility and delivery efficiency.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, Italy
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yalda Saghari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hassan Karimi-Maleh
- School of Resources and Environment, University of Electronic Science and Technology of China, Chengdu, PR China
| | | | - Esmaeel Sharifi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Türkiye
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, New South Wales, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mingzhi Shen
- Department of Cardiology, Hainan Hospital of PLA General Hospital, Sanya, China
| |
Collapse
|
18
|
Krajcer A, Grzywna E, Lewandowska-Łańcucka J. Strategies increasing the effectiveness of temozolomide at various levels of anti-GBL therapy. Biomed Pharmacother 2023; 165:115174. [PMID: 37459661 DOI: 10.1016/j.biopha.2023.115174] [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: 05/09/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/17/2023] Open
Abstract
Glioblastoma (GBL) is the most common (60-70% of primary brain tumours) and the most malignant of the glial tumours. Although current therapies remain palliative, they have been proven to prolong overall survival. Within an optimal treatment regimen (incl. surgical resection, radiation therapy, and chemotherapy) temozolomide as the current anti-GBL first-line chemotherapeutic has increased the median overall survival to 14-15 months, and the percentage of patients alive at two years has been reported to rise from 10.4% to 26.5%. Though, the effectiveness of temozolomide chemotherapy is limited by the serious systemic, dose-related side effects. Therefore, the ponderation regarding novel treatment methods along with innovative formulations is crucial to emerging the therapeutic potential of the widely used drug simultaneously reducing the drawbacks of its use. Herein the complex temozolomide application restrictions present at different levels of therapy as well as, the currently proposed strategies aimed at reducing those limitations are demonstrated. Approaches increasing the efficacy of anti-GBL treatment are addressed. Our paper is focused on the most recent developments in the field of nano/biomaterials-based systems for temozolomide delivery and their functionalization towards more effective blood-brain-barrier crossing and/or tumour targeting. Appropriate designing accounting for the physical and chemical features of formulations along with distinct routes of administration is also discussed. In addition, considering the multiple resistance mechanisms, the molecular heterogeneity and the evolution of tumour the purposely selected delivery methods, the combined therapeutic approaches and specifically focused on GBL cells therapies are reviewed.
Collapse
Affiliation(s)
- Aleksandra Krajcer
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Ewelina Grzywna
- Department of Neurosurgery and Neurotraumatology, Jagiellonian University Medical College, Św. Anny 12, 31-008 Kraków, Poland
| | | |
Collapse
|
19
|
Sharda D, Choudhury D. Insulin-cobalt core-shell nanoparticles for receptor-targeted bioimaging and diabetic wound healing. RSC Adv 2023; 13:20321-20335. [PMID: 37425626 PMCID: PMC10323873 DOI: 10.1039/d3ra01473h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/28/2023] [Indexed: 07/11/2023] Open
Abstract
Diabetic wounds represent a major issue in medical care and need advanced therapeutic and tissue imaging systems for better management. The utilization of nano-formulations involving proteins like insulin and metal ions plays significant roles in controlling wound outcomes by decreasing inflammation or reducing microbial load. This work reports the easy one-pot synthesis of extremely stable, biocompatible, and highly fluorescent insulin-cobalt core-shell nanoparticles (ICoNPs) with enhanced quantum yield for their highly specific receptor-targeted bioimaging and normal and diabetic wound healing in vitro (HEKa cell line). The particles were characterized using physicochemical properties, biocompatibility, and wound healing applications. FTIR bands at 670.35 cm-1, 849.79, and 973.73 indicating the Co-O bending, CoO-OH bond, and Co-OH bending, respectively, confirm the protein-metal interactions, which is further supported by the Raman spectra. In silico studies indicate the presence of cobalt binding sites on the insulin chain B at 8 GLY, 9 SER, and 10 HIS positions. The particles exhibit a magnificent loading efficiency of 89.48 ± 0.049% and excellent release properties (86.54 ± 2.15% within 24 h). Further, based on fluorescent properties, the recovery process can be monitored under an appropriate setup, and the binding of ICoNPs to insulin receptors was confirmed by bioimaging. This work helps synthesize effective therapeutics with numerous wound-healing promoting and monitoring applications.
Collapse
Affiliation(s)
- Deepinder Sharda
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology Patiala 147004 Punjab India +91-8196949843
| | - Diptiman Choudhury
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology Patiala 147004 Punjab India +91-8196949843
- Thapar Institute of Engineering and Technology-Virginia Tech (USA) Center of Excellence in Emerging Materials, Thapar Institute of Engineering and Technology Patiala Punjab-147004 India
| |
Collapse
|
20
|
Lv J, Wang X, Zhang X, Xu R, Hu S, Wang S, Li M. Tumor microenvironment-responsive artesunate loaded Z-scheme heterostructures for synergistic photo-chemodynamic therapy of hypoxic tumor. Asian J Pharm Sci 2023; 18:100798. [PMID: 37252037 PMCID: PMC10209134 DOI: 10.1016/j.ajps.2023.100798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/15/2023] [Accepted: 02/26/2023] [Indexed: 05/31/2023] Open
Abstract
Tumor microenvironment (TME) with the particular features of severe hypoxia, insufficient endogenous H2O2, and overexpression of glutathione (GSH) markedly reduced the antitumor efficacy of monotherapy. Herein, a TME-responsive multifunctional nanoplatform (Bi2S3@Bi@PDA-HA/Art NRs) was presented for synergistic photothermal therapy (PTT), chemodynamic therapy (CDT), and photodynamic therapy (PDT) to achieve better therapeutic outcomes. The Z-scheme heterostructured bismuth sulfide@bismuth nanorods (Bi2S3@Bi NRs) guaranteed excellent photothermal performance of the nanoplatform. Moreover, its ability to produce O2 and reactive oxygen species (ROS) synchronously could relieve tumor hypoxia and improve PDT outcomes. The densely coated polydopamine/ammonium bicarbonate (PDA/ABC) and hyaluronic acid (HA) layers on the surface of the nanoplatform enhanced the cancer-targeting capacity and induced the acidic TME-triggered in situ "bomb-like" release of Art. The CDT treatment was achieved by activating the released Art through intracellular Fe2+ ions in an H2O2-independent manner. Furthermore, decreasing the glutathione peroxidase 4 (GPX4) levels by Art could also increase the PDT efficiency of Bi2S3@Bi NRs. Owing to the synergistic effect, this nanoplatform displayed improved antitumor efficacy with minimal toxicity both in vitro and in vivo. Our design sheds light on the application of phototherapy combined with the traditional Chinese medicine monomer-artesunate in treating the hypoxic tumor.
Collapse
Affiliation(s)
- Jie Lv
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
- Postdoctoral Mobile Station of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoyu Wang
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Xue Zhang
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Runpei Xu
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuyang Hu
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuangling Wang
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Meng Li
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
21
|
Tran TH, Tran PTT, Truong DH. Lactoferrin and Nanotechnology: The Potential for Cancer Treatment. Pharmaceutics 2023; 15:pharmaceutics15051362. [PMID: 37242604 DOI: 10.3390/pharmaceutics15051362] [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: 02/20/2023] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Lactoferrin (Lf)-a glycoprotein of the transferrin family-has been investigated as a promising molecule with diverse applications, including infection inhibition, anti-inflammation, antioxidant properties and immune modulation. Along with that, Lf was found to inhibit the growth of cancerous tumors. Owing to unique properties such as iron-binding and positive charge, Lf could interrupt the cancer cell membrane or influence the apoptosis pathway. In addition, being a common mammalian excretion, Lf offers is promising in terms of targeting delivery or the diagnosis of cancer. Recently, nanotechnology significantly enhanced the therapeutic index of natural glycoproteins such as Lf. Therefore, in the context of this review, the understanding of Lf is summarized and followed by different strategies of nano-preparation, including inorganic nanoparticles, lipid-based nanoparticles and polymer-based nanoparticles in cancer management. At the end of the study, the potential future applications are discussed to pave the way for translating Lf into actual usage.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Faculty of Pharmacy, Phenikaa University, Yen Nghia, Ha Dong, Hanoi 12116, Vietnam
| | - Phuong Thi Thu Tran
- Department of Life Sciences, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi 10000, Vietnam
| | | |
Collapse
|
22
|
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]
|
23
|
Proteins and their functionalization for finding therapeutic avenues in cancer: Current status and future prospective. Biochim Biophys Acta Rev Cancer 2023; 1878:188862. [PMID: 36791920 DOI: 10.1016/j.bbcan.2023.188862] [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: 06/24/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/15/2023]
Abstract
Despite the remarkable advancement in the health care sector, cancer remains the second most fatal disease globally. The existing conventional cancer treatments primarily include chemotherapy, which has been associated with little to severe side effects, and radiotherapy, which is usually expensive. To overcome these problems, target-specific nanocarriers have been explored for delivering chemo drugs. However, recent reports on using a few proteins having anticancer activity and further use of them as drug carriers have generated tremendous attention for furthering the research towards cancer therapy. Biomolecules, especially proteins, have emerged as suitable alternatives in cancer treatment due to multiple favourable properties including biocompatibility, biodegradability, and structural flexibility for easy surface functionalization. Several in vitro and in vivo studies have reported that various proteins derived from animal, plant, and bacterial species, demonstrated strong cytotoxic and antiproliferative properties against malignant cells in native and their different structural conformations. Moreover, surface tunable properties of these proteins help to bind a range of anticancer drugs and target ligands, thus making them efficient delivery agents in cancer therapy. Here, we discuss various proteins obtained from common exogenous sources and how they transform into effective anticancer agents. We also comprehensively discuss the tumor-killing mechanisms of different dietary proteins such as bovine α-lactalbumin, hen egg-white lysozyme, and their conjugates. We also articulate how protein nanostructures can be used as carriers for delivering cancer drugs and theranostics, and strategies to be adopted for improving their in vivo delivery and targeting. We further discuss the FDA-approved protein-based anticancer formulations along with those in different phases of clinical trials.
Collapse
|
24
|
Abdelgalil R, Khattab SN, Ebrahim S, Elkhodairy KA, Teleb M, Bekhit AA, Sallam MA, Elzoghby AO. Engineered Sericin-Tagged Layered Double Hydroxides for Combined Delivery of Pemetrexed and ZnO Quantum Dots as Biocompatible Cancer Nanotheranostics. ACS OMEGA 2023; 8:5655-5671. [PMID: 36816638 PMCID: PMC9933221 DOI: 10.1021/acsomega.2c07128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/20/2023] [Indexed: 05/25/2023]
Abstract
Despite extensive progress in the field of cancer nanotheranostics, clinical development of biocompatible theranostic nanomedicine remains a formidable challenge. Herein, we engineered biocompatible silk-sericin-tagged inorganic nanohybrids for efficient treatment and imaging of cancer cells. The developed nanocarriers are anticipated to overcome the premature release of the chemotherapeutic drug pemetrexed (PMX), enhance the colloidal stability of layered double hydroxides (LDHs), and maintain the luminescence properties of ZnO quantum dots (QDs). Materials and Methods: PMX-intercalated LDHs were modified with sericin and coupled to ZnO QDs for therapy and imaging of breast cancer cells. Results: The optimized nanomedicine demonstrated a sustained release profile of PMX, and high cytotoxicity against MDA-MB-231 cells compared to free PMX. In addition, high cellular uptake of the engineered nanocarriers into MDA-MB-231 breast cancer cells was accomplished. Conclusions: Conclusively, the LDH-sericin nanohybrids loaded with PMX and conjugated to ZnO QDs offered a promising cancer theranostic nanomedicine.
Collapse
Affiliation(s)
- Riham
M. Abdelgalil
- Department
of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Cancer
Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Sherine N. Khattab
- Chemistry
Department, Faculty of Science, Alexandria
University, Alexandria 21321, Egypt
| | - Shaker Ebrahim
- Department
of Materials Science, Institute of Graduate Studies and Research, Alexandria 21526, Egypt
| | - Kadria A. Elkhodairy
- Department
of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Cancer
Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mohamed Teleb
- Cancer
Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Adnan A. Bekhit
- Cancer
Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Pharmacy
Program, Allied Health Department, College of Health and Sport Sciences, University of Bahrain, Zallaq 32038, Kingdom of Bahrain
| | - Marwa A. Sallam
- Department
of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed O. Elzoghby
- Department
of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Cancer
Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Division
of Engineering in Medicine, Department of Medicine, Brigham and Women’s
Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
25
|
Rani R, Malik P, Dhania S, Mukherjee TK. Recent Advances in Mesoporous Silica Nanoparticle-Mediated Drug Delivery for Breast Cancer Treatment. Pharmaceutics 2023; 15:227. [PMID: 36678856 PMCID: PMC9860911 DOI: 10.3390/pharmaceutics15010227] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Breast cancer (BC) currently occupies the second rank in cancer-related global female deaths. Although consistent awareness and improved diagnosis have reduced mortality in recent years, late diagnosis and resistant response still limit the therapeutic efficacy of chemotherapeutic drugs (CDs), leading to relapse with consequent invasion and metastasis. Treatment with CDs is indeed well-versed but it is badly curtailed with accompanying side effects and inadequacies of site-specific drug delivery. As a result, drug carriers ensuring stealth delivery and sustained drug release with improved pharmacokinetics and biodistribution are urgently needed. Core-shell mesoporous silica nanoparticles (MSNPs) have recently been a cornerstone in this context, attributed to their high surface area, low density, robust functionalization, high drug loading capacity, size-shape-controlled functioning, and homogeneous shell architecture, enabling stealth drug delivery. Recent interest in using MSNPs as drug delivery vehicles has been due to their functionalization and size-shape-driven versatilities. With such insights, this article focuses on the preparation methods and drug delivery mechanisms of MSNPs, before discussing their emerging utility in BC treatment. The information compiled herein could consolidate the database for using inorganic nanoparticles (NPs) as BC drug delivery vehicles in terms of design, application and resolving post-therapy complications.
Collapse
Affiliation(s)
- Ruma Rani
- ICAR-National Research Centre on Equines, Hisar 125001, Haryana, India
| | - Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Sunena Dhania
- Department of Bio & Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India
| | - Tapan Kumar Mukherjee
- Institute of Biotechnology (AIB), Amity University, Noida 201313, Uttar Pradesh, India
| |
Collapse
|
26
|
AbouAitah K, Soliman AAF, Swiderska-Sroda A, Nassrallah A, Smalc-Koziorowska J, Gierlotka S, Lojkowski W. Co-Delivery System of Curcumin and Colchicine Using Functionalized Mesoporous Silica Nanoparticles Promotes Anticancer and Apoptosis Effects. Pharmaceutics 2022; 14:pharmaceutics14122770. [PMID: 36559264 PMCID: PMC9785757 DOI: 10.3390/pharmaceutics14122770] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Purpose: Many natural agents have a high anticancer potential, and their combination may be advantageous for improved anticancer effects. Such agents, however, often are not water soluble and do not efficiently target cancer cells, and the kinetics of their action is poorly controlled. One way to overcome these barriers is to combine natural agents with nanoparticles. Our aim in the current study was to fabricate an anticancer nanoformulation for co-delivery of two natural agents, curcumin (CR) and colchicine (CL), with a core-shell structure. Using cancer cell lines, we compared the anticancer efficacy between the combination and a nanoformulation with CL alone. Methods: For the single-drug nanoformulation, we used phosphonate groups to functionalize mesoporous silica nanoparticles (MSNs) and loaded the MSNs with CL. Additional loading of this nanoformulation with CR achieved the co-delivery format. To create the structure with a core shell, we selected a chitosan−cellulose mixture conjugated with targeting ligands of folic acid for the coating. For evaluating anticancer and apoptosis effects, we assessed changes in important genes and proteins in apoptosis (p53, caspase-3, Bax, Bcl-2) in several cell lines (MCF-7, breast adenocarcinoma; HCT-116, colon carcinoma; HOS, human osteosarcoma; and A-549, non−small cell lung cancer). Results: Nanoformulations were successfully synthesized and contained 10.9 wt.% for the CL single-delivery version and 18.1 wt.% for the CL+CR co-delivery nanoformulation. Anticancer effects depended on treatment, cell line, and concentration. Co-delivery nanoformulations exerted anticancer effects that were significantly superior to those of single delivery or free CL or CR. Anticancer effects by cell line were in the order of HCT-116 > A549 > HOS > MCF-7. The lowest IC50 value was obtained for the nanoformulation consisting of CL and CR coated with a polymeric shell conjugated with FA (equivalent to 4.1 ± 0.05 µg/mL). With dual delivery compared with the free agents, we detected strongly increased p53, caspase-3, and Bax expression, but inhibition of Bcl-2, suggesting promotion of apoptosis. Conclusions: Our findings, although preliminary, indicate that the proposed dual delivery nanoformulation consisting of nanocore: MSNs loaded with CL and CR and coated with a shell of chitosan−cellulose conjugated folic acid exerted strong anticancer and apoptotic effects with potent antitumor activity against HCT-116 colon cells. The effect bested CL alone. Evaluating and confirming the efficacy of co-delivery nanoformulations will require in vivo studies.
Collapse
Affiliation(s)
- Khaled AbouAitah
- Medicinal and Aromatic Plants Research Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC), 33 El-Behouth Street, Dokki, Giza 12622, Egypt
- Correspondence: (K.A.); (W.L.); Tel.: +20-233371635 (K.A.); +48-22-888-0429 or +48-22-632-4302 (W.L.); Fax: +20-233371010 (K.A.); +48-22-632-4218 (W.L.)
| | - Ahmed A. F. Soliman
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC), 33 El-Behouth St, Dokki, Giza 12622, Egypt
| | - Anna Swiderska-Sroda
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland
| | - Amr Nassrallah
- Biochemistry Department, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| | - Julita Smalc-Koziorowska
- Laboratory of Semiconductor Characterization, Institute of High Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland
| | - Stanislaw Gierlotka
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland
| | - Witold Lojkowski
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland
- Correspondence: (K.A.); (W.L.); Tel.: +20-233371635 (K.A.); +48-22-888-0429 or +48-22-632-4302 (W.L.); Fax: +20-233371010 (K.A.); +48-22-632-4218 (W.L.)
| |
Collapse
|
27
|
Deepika, Maurya PK. Ellagic acid: insight into its protective effects in age-associated disorders. 3 Biotech 2022; 12:340. [PMID: 36340805 PMCID: PMC9633905 DOI: 10.1007/s13205-022-03409-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
The disparity in the free radical generation and the production of antioxidants to counteract its effect is known as oxidative stress. Oxidative stress causes damage to the macromolecules such as lipids, carbohydrates, proteins, and DNA and RNA. The oxidative damage to the cellular components leads to a process of aging and various age-associated disorders. The literature survey for this review was done using PubMed, Google Scholar, and Science Direct. The papers showing the studies related to aging and age-associated disorders have been selected for reviewing this paper. Ellagic acid has been used as the keyword, and more emphasis has been put on papers from the last 10 years. However, some papers with significant studies prior to 10 years have also been considered. Almost 250 papers have been studied for reviewing this paper, and about 135 papers have been cited. Ellagic acid (EA) is present in high quantities in pomegranate and various types of berries. It is known to possess the antioxidant potential and protects from the harmful effects of free radicals. Various studies have shown its effect to protect cardiovascular, neurodegenerative, cancer, and diabetes. The present review focuses on the protective effect of ellagic acid in age-associated disorders. The effect of EA has been studied in various chronic disorders but the scope of this review is limited to cancer, diabetes, cardiovascular and neurodegenerative disorders. All the disease aspects have not been addressed in this particular review.
Collapse
Affiliation(s)
- Deepika
- Department of Biochemistry, Central University of Haryana, Mahendragarh, 123031 India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendragarh, 123031 India
| |
Collapse
|
28
|
Mohammadinejad A, Mohajeri T, Aleyaghoob G, Heidarian F, Kazemi Oskuee R. Ellagic acid as a potent anticancer drug: A comprehensive review on in vitro, in vivo, in silico, and drug delivery studies. Biotechnol Appl Biochem 2022; 69:2323-2356. [PMID: 34846078 DOI: 10.1002/bab.2288] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 11/10/2021] [Indexed: 12/27/2022]
Abstract
Ellagic acid as a polyphenol or micronutrient, which can be naturally found in different vegetables and fruits, has gained considerable attention for cancer therapy due to considerable biological activities and different molecular targets. Ellagic acid with low hydrolysis and lipophilic and hydrophobic nature is not able to be absorbed in circulation. So, accumulation inside the intestinal epithelial cells or metabolization to other urolithins leads to the limitation of direct evaluation of EA effects in clinical studies. This review focuses on the studies which supported anticancer activity of pure or fruit-extracted ellagic acid through in vitro, in vivo, in silico, and drug delivery methods. The results demonstrate ellagic acid modulates the expression of various genes incorporated in the cancer-related process of apoptosis and proliferation, inflammation related-gens, and oxidative-related genes. Moreover, the ellagic acid formulation in carriers composed of lipid, silica, chitosan, iron- bovine serum albumin nanoparticles obviously enhanced the stable release and confident delivery with minimum loss. Also, in silico analysis proved that ellagic acid was able to be placed at a position of cocrystal ADP, in the deep cavity of the protein target, and tightly interact with binding pocket residues leading to suppression of substrate availability of protein and its activation inhibition.
Collapse
Affiliation(s)
- Arash Mohammadinejad
- Targeted Drug Delivery Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Taraneh Mohajeri
- Department of Obstetrics & Gynecology, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran
| | - Ghazaleh Aleyaghoob
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Heidarian
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
29
|
Wang Y, Zhu M, Shi T, Ma X, Wu X, Li QX, Hua R. Construction of a novel fluorescent nanocarrier with double hollow shells for pH-controlled release of imidacloprid and its distribution and transport in bok choy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 246:114132. [PMID: 36206638 DOI: 10.1016/j.ecoenv.2022.114132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
Nanotechnology has been widely used in the field of pesticides. Integration of nano-pesticides and carbon dot fluorescence can fully utilize the potential for high admission of pesticides on leaves and convenience observation of its distribution and transport in the tissues. In the present study, a fluorescent mesoporous nanosilica with double hollow shells for loading imidacloprid (Im@FL-MSNs) was designed and synthesized. The physical and chemical properties of the imidacloprid nanocarriers were characterized by transmission electron microscopy (TEM), FT-IR spectroscopy, thermogravimetric analysis (TGA), X-ray photoelectron spectroscopy (XPS) and N2 adsorption/desorption. When the mass ratio of FL MSNs to imidacloprid is 6:5, Im@FL-MSNs exhibits good fluorescence properties, high loading efficiency (∼30%), great slow-release performance as well as pH controllability. Besides, Im@FL-MSNs can improve the ability of imidacloprid to adhere on the leaf surface of bok choy (Initial contact angled is greater than 80°). Importantly, Im@FL-MSNs did not reduce the biological activity of imidacloprid (LC50 (95% CI) = 1.43 mg/L). It was able to visually study the absorption and distribution of imidacloprid in bok choy plants, and provide theoretical and technical guidance for pesticide reduction.
Collapse
Affiliation(s)
- Yi Wang
- Key Laboratory of Agri-Food Safety of Anhui Province, Department of Pesticide Science, School of Resources and Environment, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei 230036, China
| | - Meiqing Zhu
- Key Laboratory of Agri-Food Safety of Anhui Province, Department of Pesticide Science, School of Resources and Environment, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei 230036, China; School of Chemical and Environmental Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Taozhong Shi
- Key Laboratory of Agri-Food Safety of Anhui Province, Department of Pesticide Science, School of Resources and Environment, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei 230036, China
| | - Xin Ma
- Key Laboratory of Agri-Food Safety of Anhui Province, Department of Pesticide Science, School of Resources and Environment, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei 230036, China
| | - Xiangwei Wu
- Key Laboratory of Agri-Food Safety of Anhui Province, Department of Pesticide Science, School of Resources and Environment, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei 230036, China
| | - Qing X Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, 1955 East-West Road, Honolulu, HI 96822, USA
| | - Rimao Hua
- Key Laboratory of Agri-Food Safety of Anhui Province, Department of Pesticide Science, School of Resources and Environment, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei 230036, China.
| |
Collapse
|
30
|
R. M. Metawea O, Teleb M, Haiba NS, Elzoghby AO, Khafaga AF, Noreldin AE, Khattab SN, Khalil HH. Folic acid-poly(N-isopropylacrylamide-maltodextrin) nanohydrogels a novel thermo-/pH-responsive polymer for resveratrol breast cancer targeted therapy. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
A comparative evaluation of anti-tumor activity following oral and intravenous delivery of doxorubicin in a xenograft model of breast tumor. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00595-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose
Natural materials have been extensively studied for oral drug delivery due to their biodegradability and other unique properties. In the current research, we fabricated sodium caseinate nanomicelles (NaCNs) using casein as a natural polymer to develop a controlled-release oral delivery system that would improve the therapeutic potential of doxorubicin (DOX) and reduce its toxicity.
Methods
DOX-loaded NaCNs were synthesized and thoroughly characterized, then subjected to in vivo anti-tumor evaluation and bio-distribution analysis in a 4T1-induced breast cancer model.
Results
Our findings indicated that the tumor would shrink by eight-fold in the group orally treated with DOX-NaCNs when compared to free DOX. The tumor accumulated drug 1.27-fold more from the orally administered DOX-NaCNs compared to the intravenously administered DOX-NaCNs, 6.8-fold more compared to free DOX, and 8.34-times more compared to orally administered free DOX. In comparison, the orally administered DOX-NaCNs lead to a significant reduction in tumor size (5.66 ± 4.36 mm3) compared to intravenously administered DOX-NaCNs (10.29 ± 4.86 mm3) on day 17 of the experiment. NaCNs were well tolerated at a single dose of 2000 mg/kg in an acute oral toxicity study.
Conclusion
The enhanced anti-tumor effects of oral DOX-NaCNs might be related to the controlled release of DOX from the delivery system when compared to free DOX and the intravenous formulation of DOX-NaCNs. Moreover, NaCNs is recognized as a safe and non-toxic delivery system with excellent bio-distribution profile and high anti-tumor effects that has a potential for oral chemotherapy.
Collapse
|
32
|
Krzyzowska M, Janicka M, Tomaszewska E, Ranoszek-Soliwoda K, Celichowski G, Grobelny J, Szymanski P. Lactoferrin-Conjugated Nanoparticles as New Antivirals. Pharmaceutics 2022; 14:pharmaceutics14091862. [PMID: 36145610 PMCID: PMC9504495 DOI: 10.3390/pharmaceutics14091862] [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: 08/01/2022] [Revised: 08/20/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Lactoferrin is an iron-binding glycoprotein with multiple functions in the body. Its activity against a broad spectrum of both DNA and RNA viruses as well as the ability to modulate immune responses have made it of interest in the pharmaceutical and food industries. The mechanisms of its antiviral activity include direct binding to the viruses or its receptors or the upregulation of antiviral responses by the immune system. Recently, much effort has been devoted to the use of nanotechnology in the development of new antivirals. In this review, we focus on describing the antiviral mechanisms of lactoferrin and the possible use of nanotechnology to construct safe and effective new antiviral drugs.
Collapse
Affiliation(s)
- Malgorzata Krzyzowska
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
- Correspondence:
| | - Martyna Janicka
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Emilia Tomaszewska
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Katarzyna Ranoszek-Soliwoda
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Grzegorz Celichowski
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Jarosław Grobelny
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Pawel Szymanski
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| |
Collapse
|
33
|
Sargazi S, Laraib U, Barani M, Rahdar A, Fatima I, Bilal M, Pandey S, Sharma RK, Kyzas GZ. Recent trends in mesoporous silica nanoparticles of rode-like morphology for cancer theranostics: A review. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
34
|
Li Y, Deng G, Hu X, Li C, Wang X, Zhu Q, Zheng K, Xiong W, Wu H. Recent advances in mesoporous silica nanoparticle-based targeted drug-delivery systems for cancer therapy. Nanomedicine (Lond) 2022; 17:1253-1279. [PMID: 36250937 DOI: 10.2217/nnm-2022-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Targeted drug-delivery systems are a growing research topic in tumor treatment. In recent years, mesoporous silica nanoparticles (MSNs) have been extensively studied and applied in noninvasive and biocompatible drug-delivery systems for tumor therapy due to their outstanding advantages, which include high surface area, large pore volume, tunable pore size, easy surface modification and stable framework. The advances in the application of MSNs for anticancer drug targeting are covered and highlighted in this review, and the challenges and prospects of MSN-based targeted drug-delivery systems are discussed. This review provides new insights for researchers interested in targeted drug-delivery systems against cancer.
Collapse
Affiliation(s)
- Ying Li
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Guoxing Deng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China.,School of Pharmacy, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Xianlong Hu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Chenyang Li
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Xiaodong Wang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Qinchang Zhu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Kai Zheng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Wei Xiong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Haiqiang Wu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| |
Collapse
|
35
|
Ahmadi A, Sokunbi M, Patel T, Chang MW, Ahmad Z, Singh N. Influence of Critical Parameters on Cytotoxicity Induced by Mesoporous Silica Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2016. [PMID: 35745355 PMCID: PMC9228019 DOI: 10.3390/nano12122016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 02/01/2023]
Abstract
Mesoporous Silica Nanoparticles (MSNs) have received increasing attention in biomedical applications due to their tuneable pore size, surface area, size, surface chemistry, and thermal stability. The biocompatibility of MSNs, although generally believed to be satisfactory, is unclear. Physicochemical properties of MSNs, such as diameter size, morphology, and surface charge, control their biological interactions and toxicity. Experimental conditions also play an essential role in influencing toxicological results. Therefore, the present study includes studies from the last five years to statistically analyse the effect of various physicochemical features on MSN-induced in-vitro cytotoxicity profiles. Due to non-normally distributed data and the presence of outliers, a Kruskal-Wallis H test was conducted on different physicochemical characteristics, including diameter sizes, zeta-potential measurements, and functionalisation of MSNs, based on the viability results, and statistical differences were obtained. Subsequently, pairwise comparisons were performed using Dunn's procedure with a Bonferroni correction for multiple comparisons. Other experimental parameters, such as type of cell line used, cell viability measurement assay, and incubation time, were also explored and analysed for statistically significant results.
Collapse
Affiliation(s)
- Amirsadra Ahmadi
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (A.A.); (M.S.); (T.P.)
| | - Moses Sokunbi
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (A.A.); (M.S.); (T.P.)
| | - Trisha Patel
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (A.A.); (M.S.); (T.P.)
| | - Ming-Wei Chang
- Nanotechnology and Integrated Bioengineering Centre, Jordanstown Campus, University of Ulster, Newtownabbey BT37 0QB, UK;
| | - Zeeshan Ahmad
- Leicester School of Pharmaceutical Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK;
| | - Neenu Singh
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (A.A.); (M.S.); (T.P.)
| |
Collapse
|
36
|
Green Self-assembled Lactoferrin Carboxymethyl Cellulose Nanogels for Synergistic Chemo/herbal Breast Cancer Therapy. Colloids Surf B Biointerfaces 2022; 217:112657. [DOI: 10.1016/j.colsurfb.2022.112657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 11/18/2022]
|
37
|
The Role of the NMDA Receptor in the Anticonvulsant Effect of Ellagic Acid in Pentylenetetrazole-Induced Seizures in Male Mice. Behav Neurol 2022; 2022:9015842. [PMID: 35600241 PMCID: PMC9117013 DOI: 10.1155/2022/9015842] [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: 04/24/2021] [Revised: 02/15/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Methods In this experimental study, 64 mice were divided into 8 groups and received the following: normal saline; EA at doses of 6.25, 12.5, and 25 mg/kg; NMDA agonist at a dose of 75 mg/kg; NMDA antagonist (ketamine) at a dose of 0.5 mg/kg; an effective dose of EA plus NMDA agonist; and a subeffective dose of EA plus ketamine. We induced seizure using intravenous administration of PTZ. 60 minutes before induction of seizure, drugs were administrated. Duration lasts to seizure-induced was measured. Finally, the gene expression of NMDA receptor subunits (Nr2a and Nr2b) was assessed in the prefrontal cortex. Results Results showed that EA increased the seizure threshold and decreased the expression of Nr2a and Nr2b. We determined that ketamine potentiated and NMDA attenuated the effects of subeffective and effective doses of EA. Conclusion EA probably via attenuation of the NMDA-R pathway possesses an anticonvulsant effect in PTZ-induced seizure in mice.
Collapse
|
38
|
Konhäuser M, Kannaujiya VK, Steiert E, Schwickert K, Schirmeister T, Wich PR. Co-Encapsulation of l-Asparaginase and Etoposide in Dextran Nanoparticles for Synergistic Effect in Chronic Myeloid Leukemia Cells. Int J Pharm 2022; 622:121796. [PMID: 35525474 DOI: 10.1016/j.ijpharm.2022.121796] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/02/2022] [Accepted: 04/29/2022] [Indexed: 11/17/2022]
Abstract
Co-encapsulation of multiple therapeutic drugs in a single nanocarrier has the potential to enable synergistic interactions, increase drug efficacy, and reduce side effects. The enzyme l-asparaginase and the small molecule drug etoposide have a known synergistic effect against selected cancer types. However, both drugs differ significantly in size, molecular weight, and solubility, which often results in challenges when a simultaneous delivery is required. In this study, we present the co-encapsulation of a large hydrophilic enzyme l-asparaginase and the small hydrophobic drug etoposide into a biodegradable, biocompatible, and acid-responsive dextran-based nanoparticle system. These dual drug-loaded nanoparticles show an excellent cellular uptake in chronic myeloid leukemia (CML) K562 cells and a stepwise release of the cytotoxic payloads in a pH-dependent manner. In activity tests, the dual drug-loaded formulation has shown a significant effect on cell viability (down to 31%) compared to those incubated only with l-asparaginase (92%) or etoposide (82%) at a particle concentration of 125 μg∙mL-1. These results show that the simultaneous co-delivery of these two drugs in K562 cells leads to synergistic cytotoxicity, indicating a great potential for the treatment of CML.
Collapse
Affiliation(s)
- M Konhäuser
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - V K Kannaujiya
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - E Steiert
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - K Schwickert
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - T Schirmeister
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - P R Wich
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany; School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
39
|
Xue P, Zhang G, Zhang J, Ren L. Synergism of ellagic acid in combination with radiotherapy and chemotherapy for cancer treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153998. [PMID: 35217437 DOI: 10.1016/j.phymed.2022.153998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Ellagic acid (EA) is a polyphenol compound abundant in berries, walnuts, pecans, pomegranate, cranberries, and other plant foods and exerts a wide array of biological properties. In particular, EA has received considerable research attention in anti-cancer therapy. EA administered alone has been shown to exert effects against human cancers through multiple pathways. In addition, EA may increase tumor sensitivity to chemotherapy and radiotherapy. Namely, EA combination with a relatively low dosage of therapeutic drugs or optimized radiation dose could improve the treatment outcome. More importantly, EA could counteract chemotherapy-related adverse reactions. PURPOSE This review aims to summarize the in vitro and in vivo experimental evidence of synergism of EA in radiotherapy/chemotherapy for the treatment of cancers. In addition, the preventive effect of EA to counteract chemotherapy-induced toxicity is also discussed. METHODS The searches were performed in the PubMed, Web of Science and Google scholar and introduced the information about the role of EA in cancer treatment. RESULTS EA exhibits synergistic effects in radiotherapy/chemotherapy for the treatment of cancers and exerts a great potential in reducing the side effects of chemotherapy and radiotherapy due to its biological activities, such as antioxidant and anti-inflammatory activities. CONCLUSION EA could be a promising drug adjuvant for cancer treatment. In the near future, novel strategies for EA delivery systems that overcome the low EA solubility and bioavailability should be studied further to fully exploit the therapeutic potential of EA.
Collapse
Affiliation(s)
- Peiyu Xue
- College of Food Science and Engineering, Jilin University, Changchun 130062, China; School of Biology and Food Engineering, Anyang Institute of Technology, Anyang 455000, China
| | - Guangjie Zhang
- School of Biology and Food Engineering, Anyang Institute of Technology, Anyang 455000, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
40
|
Ding Q, Xu Z, Zhou L, Rao C, Li W, Muddassir M, Sakiyama H, Li B, Ouyang Q, Liu J. A multimodal Metal-Organic framework based on unsaturated metal site for enhancing antitumor cytotoxicity through Chemo-Photodynamic therapy. J Colloid Interface Sci 2022; 621:180-194. [PMID: 35461133 DOI: 10.1016/j.jcis.2022.04.078] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023]
Abstract
Chemodynamic therapy when combined with chemotherapy opens up a new avenue for treatment of cancer. However, its development is still restricted by low targeting, high dose and toxic side effects. Herein, rational designing and construction of a new multifunctional platform with the core-shell structure 5-ALA@UiO-66-NH-FAM@CP1 (ALA = 5-aminolevulinic acid, CP1 = zirconium-pemetrexed (Zr-MTA)) has been performed. In this platform, CP1 acting as a shell is encapsulated with the UiO-66-NH2 to engender a core-shell structure that promotes and achieves a high MTA loading rate through high affinity between MTA and unsaturated Zr site of UiO-66-NH2. The 5-ALA and 5-carboxyl fluorescein (5-FAM) was successfully loaded and covalently combined with UiO-66-NH2 due to its high porosity and presence of amino groups. The characterization results indicated that the loading rate of MTA (41.03 wt%) of platform is higher than the reported values. More importantly, the in vitro and in vivo results also demonstrated that it has a good folate targeting ability and realizes high efficient antitumor activity by chemotherapy combied with photodynamic therapy (PDT). This newly developed multifunctional platform could provide a new idea for designing and constructing the carrier with chemotherapy and PDT therapy.
Collapse
Affiliation(s)
- Qiongjie Ding
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan China; Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Zhijue Xu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Luyi Zhou
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Congying Rao
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China
| | - Weimin Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mohd Muddassir
- Department of Chemistry, College of Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hiroshi Sakiyama
- Department of Science, Faculty of Science, Yamagata University, 1-4-12 Kojirakawa, Yamagata 990-8560, Japan
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Qin Ouyang
- Department of General Surgery, Dalang Hospital, Dongguan 523800, China.
| | - Jianqiang Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan China; Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan 523808, China.
| |
Collapse
|
41
|
Mokhtar S, Khattab SN, Elkhodairy KA, Teleb M, Bekhit AA, Elzoghby AO, Sallam MA. Methotrexate-Lactoferrin Targeted Exemestane Cubosomes for Synergistic Breast Cancer Therapy. Front Chem 2022; 10:847573. [PMID: 35392419 PMCID: PMC8980280 DOI: 10.3389/fchem.2022.847573] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/16/2022] [Indexed: 01/01/2023] Open
Abstract
While the treatment regimen of certain types of breast cancer involves a combination of hormonal therapy and chemotherapy, the outcomes are limited due to the difference in the pharmacokinetics of both treatment agents that hinders their simultaneous and selective delivery to the cancer cells. Herein, we report a hybrid carrier system for the simultaneous targeted delivery of aromatase inhibitor exemestane (EXE) and methotrexate (MTX). EXE was physically loaded within liquid crystalline nanoparticles (LCNPs), while MTX was chemically conjugated to lactoferrin (Lf) by carbodiimide reaction. The anionic EXE-loaded LCNPs were then coated by the cationic MTX–Lf conjugate via electrostatic interactions. The Lf-targeted dual drug-loaded LCNPs exhibited a particle size of 143.6 ± 3.24 nm with a polydispersity index of 0.180. It showed excellent drug loading with an EXE encapsulation efficiency of 95% and an MTX conjugation efficiency of 33.33%. EXE and MTX showed synergistic effect against the MCF-7 breast cancer cell line with a combination index (CI) of 0.342. Furthermore, the Lf-targeted dual drug-loaded LCNPs demonstrated superior synergistic cytotoxic activity with a combination index (CI) of 0.242 and a dose reduction index (DRI) of 34.14 and 4.7 for EXE and MTX, respectively. Cellular uptake studies demonstrated higher cellular uptake of Lf-targeted LCNPs into MCF-7 cancer cells than non-targeted LCNPs after 4 and 24 h. Collectively, the targeted dual drug-loaded LCNPs are a promising candidate offering combinational hormonal therapy/chemotherapy for breast cancer.
Collapse
Affiliation(s)
- Sarah Mokhtar
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Sherine N. Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
- *Correspondence: Sherine N. Khattab, , ; Ahmed O. Elzoghby,
| | - Kadria A. Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mohamed Teleb
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Adnan A. Bekhit
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Pharmacy Program, Allied Health Department, College of Health and Sport Sciences, University of Bahrain, Al-Manamah, Bahrain
| | - Ahmed O. Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- *Correspondence: Sherine N. Khattab, , ; Ahmed O. Elzoghby,
| | - Marwa A. Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
42
|
Kankala RK, Han YH, Xia HY, Wang SB, Chen AZ. Nanoarchitectured prototypes of mesoporous silica nanoparticles for innovative biomedical applications. J Nanobiotechnology 2022; 20:126. [PMID: 35279150 PMCID: PMC8917689 DOI: 10.1186/s12951-022-01315-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
Despite exceptional morphological and physicochemical attributes, mesoporous silica nanoparticles (MSNs) are often employed as carriers or vectors. Moreover, these conventional MSNs often suffer from various limitations in biomedicine, such as reduced drug encapsulation efficacy, deprived compatibility, and poor degradability, resulting in poor therapeutic outcomes. To address these limitations, several modifications have been corroborated to fabricating hierarchically-engineered MSNs in terms of tuning the pore sizes, modifying the surfaces, and engineering of siliceous networks. Interestingly, the further advancements of engineered MSNs lead to the generation of highly complex and nature-mimicking structures, such as Janus-type, multi-podal, and flower-like architectures, as well as streamlined tadpole-like nanomotors. In this review, we present explicit discussions relevant to these advanced hierarchical architectures in different fields of biomedicine, including drug delivery, bioimaging, tissue engineering, and miscellaneous applications, such as photoluminescence, artificial enzymes, peptide enrichment, DNA detection, and biosensing, among others. Initially, we give a brief overview of diverse, innovative stimuli-responsive (pH, light, ultrasound, and thermos)- and targeted drug delivery strategies, along with discussions on recent advancements in cancer immune therapy and applicability of advanced MSNs in other ailments related to cardiac, vascular, and nervous systems, as well as diabetes. Then, we provide initiatives taken so far in clinical translation of various silica-based materials and their scope towards clinical translation. Finally, we summarize the review with interesting perspectives on lessons learned in exploring the biomedical applications of advanced MSNs and further requirements to be explored.
Collapse
Affiliation(s)
- Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China.
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China.
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People's Republic of China.
| | - Ya-Hui Han
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
| | - Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People's Republic of China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian, People's Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, 361021, Fujian, People's Republic of China
| |
Collapse
|
43
|
Raikwar S, Jain A, Saraf S, Bidla PD, Panda PK, Tiwari A, Verma A, Jain SK. Opportunities in combinational chemo-immunotherapy for breast cancer using nanotechnology: an emerging landscape. Expert Opin Drug Deliv 2022; 19:247-268. [PMID: 35184620 DOI: 10.1080/17425247.2022.2044785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 02/17/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Breast carcinoma (BC) is one of the most frequent causes of cancer-related death among women, which is due to the poor response to conventional therapy. There are several complications associated with monotherapy for cancer, such as cytotoxicity to normal cells, multidrug resistance (MDR), side effects, and limited applications. To overcome these challenges, a combination of chemotherapy and immunotherapy (monoclonal antibodies, anticancer vaccines, checkpoint inhibitors, and cytokines) has been introduced. Drug delivery systems (DDSs) based on nanotechnology have more applications in BC treatment owing to their controlled and targeted drug release with lower toxicity and reduced adverse drug effects. Several nanocarriers, such as liposomes, nanoparticles, dendrimers, and micelles, have been used for the effective delivery of drugs. AREAS COVERED This article presents opportunities and challenges in BC treatment, the rationale for cancer immunotherapy, and several combinational approaches with their applications for BC treatment. EXPERT OPINION Nanotechnology can be used for the early prognosis and cure of BC. Several novel and targeted DDSs have been developed to enhance the efficacy of anticancer drugs. This article aims to understand new strategies for the treatment of BC and the appropriate design of nanocarriers used as a combinational DDS.
Collapse
Affiliation(s)
- Sarjana Raikwar
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Ankit Jain
- Department of Materials Engineering, Indian Institute of Science, Bangalore, India
| | - Shivani Saraf
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Pooja Das Bidla
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Pritish Kumar Panda
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Ankita Tiwari
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Amit Verma
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Pharmaceutics Research Projects Laboratory, Sagar, Madhya Pradesh, India
| |
Collapse
|
44
|
Wang S, Lv J, Pang Y, Hu S, Lin Y, Li M. Ion channel-targeting near-infrared photothermal switch with synergistic effect for specific cancer therapy. J Mater Chem B 2022; 10:748-756. [PMID: 35022632 DOI: 10.1039/d1tb02351a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite significant achievement in chemotherapy, the off-target actions and low pharmaceutical selectivity of the therapeutic agents still limit their clinical efficacy. Herein, a multifunctional nanoplatform which integrates chemotherapy, chemodynamic therapy (CDT) and photoactivation of TRPV1 channels has been successfully established for specific cancer therapy. Polydopamine (PDA) coated hollow prussian blue nanocages (hPBNCs) are used as the photothermal switches and drug carriers for loading chemotherapeutic drug, doxorubicin (Dox). Conjugating with the TRPV1 antibodies enables the nanoplatform to bind specifically to TRPV1 channels on the plasma membrane of the TRPV1-positive cancer cells and then activate them by local heating upon NIR irradiation, leading to the over-influx of Ca2+. Critically, the laser irradiation can be carefully controlled to not only open the TRPV1 channels but also avoid burning of tumors by hyperthermia. Moreover, the exposed hPBNCs in the acidic tumor cells can decompose endogenous H2O2 into ˙OH by Fenton reaction to realize CDT, which further aggravates cancer cell apoptosis. Together with the chemotherapy caused by Dox, our nanoplatform displays an enhanced anticancer effect both in vitro and in vivo. Our work provides a powerful means for site-specific cancer synergetic therapy with high spatial and temporal resolution.
Collapse
Affiliation(s)
- Shuangling Wang
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Jie Lv
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Yu Pang
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Shuyang Hu
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Yulong Lin
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Meng Li
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
45
|
Lv J, Wang S, Qiao D, Lin Y, Hu S, Li M. Mitochondria-targeting multifunctional nanoplatform for cascade phototherapy and hypoxia-activated chemotherapy. J Nanobiotechnology 2022; 20:42. [PMID: 35062959 PMCID: PMC8780403 DOI: 10.1186/s12951-022-01244-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/04/2022] [Indexed: 12/11/2022] Open
Abstract
Despite considerable progress has been achieved in hypoxia-associated anti-tumor therapy, the efficacy of utilizing hypoxia-activated prodrugs alone is not satisfied owing to the inadequate hypoxia within the tumor regions. In this work, a mitochondrial targeted nanoplatform integrating photodynamic therapy, photothermal therapy and hypoxia-activated chemotherapy has been developed to synergistically treat cancer and maximize the therapeutic window. Polydopamine coated hollow copper sulfide nanoparticles were used as the photothermal nanoagents and thermosensitive drug carriers for loading the hypoxia-activated prodrug, TH302, in our study. Chlorin e6 (Ce6) and triphenyl phosphonium (TPP) were conjugated onto the surface of the nanoplatform. Under the action of TPP, the obtained nanoplatform preferentially accumulated in mitochondria to restore the drug activity and avoid drug resistance. Using 660 nm laser to excite Ce6 can generate ROS and simultaneously exacerbate the cellular hypoxia. While under the irradiation of 808 nm laser, the nanoplatform produced local heat which can increase the release of TH302 in tumor cells, ablate cancer cells as well as intensify the tumor hypoxia levels. The aggravated tumor hypoxia then significantly boosted the anti-tumor efficiency of TH302. Both in vitro and in vivo studies demonstrated the greatly improved anti-cancer activity compared to conventional hypoxia-associated chemotherapy. This work highlights the potential of using a combination of hypoxia-activated prodrugs plus phototherapy for synergistic cancer treatment.
Collapse
Affiliation(s)
- Jie Lv
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuangling Wang
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China
| | - Duo Qiao
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yulong Lin
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuyang Hu
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China
| | - Meng Li
- College of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
46
|
Abad I, Conesa C, Sánchez L. Development of Encapsulation Strategies and Composite Edible Films to Maintain Lactoferrin Bioactivity: A Review. MATERIALS 2021; 14:ma14237358. [PMID: 34885510 PMCID: PMC8658689 DOI: 10.3390/ma14237358] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022]
Abstract
Lactoferrin (LF) is a whey protein with various and valuable biological activities. For this reason, LF has been used as a supplement in formula milk and functional products. However, it must be considered that the properties of LF can be affected by technological treatments and gastrointestinal conditions. In this article, we have revised the literature published on the research done during the last decades on the development of various technologies, such as encapsulation or composite materials, to protect LF and avoid its degradation. Multiple compounds can be used to conduct this protective function, such as proteins, including those from milk, or polysaccharides, like alginate or chitosan. Furthermore, LF can be used as a component in complexes, nanoparticles, hydrogels and emulsions, to encapsulate, protect and deliver other bioactive compounds, such as essential oils or probiotics. Additionally, LF can be part of systems to deliver drugs or to apply certain therapies to target cells expressing LF receptors. These systems also allow improving the detection of gliomas and have also been used for treating some pathologies, such as different types of tumours. Finally, the application of LF in edible and active films can be effective against some contaminants and limit the increase of the natural microbiota present in meat, for example, becoming one of the most interesting research topics in food technology.
Collapse
Affiliation(s)
- Inés Abad
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain; (I.A.); (C.C.)
- Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza-CITA, 50013 Zaragoza, Spain
| | - Celia Conesa
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain; (I.A.); (C.C.)
| | - Lourdes Sánchez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain; (I.A.); (C.C.)
- Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza-CITA, 50013 Zaragoza, Spain
- Correspondence: ; Tel.: +34-976-761-585
| |
Collapse
|
47
|
Hafez DA, Hassanin IA, Teleb M, Khattab SN, Elkhodairy KA, Elzoghby AO. Recent advances in nanomedicine-based delivery of histone deacetylase inhibitors for cancer therapy. Nanomedicine (Lond) 2021; 16:2305-2325. [PMID: 34551585 DOI: 10.2217/nnm-2021-0196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) are cancer therapeutics that operate at the epigenetic level and which have recently gained wide attention. However, the applications of HDACi are generally hindered by their poor physicochemical characteristics and unfavorable pharmacokinetic profile. Inspired by the approved nanomedicine-based drugs in the market, nanocarriers could provide a resort to circumvent the limitations imposed by HDACi. Enhanced tumor targeting, improved cellular uptake and reduced toxicity are major advantages offered by HDACi-loaded nanoparticles. More importantly, site-specific drug delivery can be achieved via engineered stimuli-responsive nanosystems. In this review we elucidate the anticancer mechanisms of HDACi and their structure-activity relationships, with a special focus on their nanomedicine-based delivery, different drug loading concepts and their implications.
Collapse
Affiliation(s)
- Dina A Hafez
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Islam A Hassanin
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Biotechnology, Institute of Graduate Studies & Research, Alexandria University, Alexandria, 21526, Egypt
| | - Mohamed Teleb
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Sherine N Khattab
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Chemistry, Faculty of Science, Alexandria University, Alexandria, 21321, Egypt
| | - Kadria A Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
48
|
Anwar DM, El-Sayed M, Reda A, Fang JY, Khattab SN, Elzoghby AO. Recent advances in herbal combination nanomedicine for cancer: delivery technology and therapeutic outcomes. Expert Opin Drug Deliv 2021; 18:1609-1625. [PMID: 34254868 DOI: 10.1080/17425247.2021.1955853] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The use of herbal compounds in cancer therapy has great potential to promote the efficacy of current cancer therapeutic strategies. Herbal compounds were successfully reported to enhance tumor cells sensitization to the action of chemo-, hormonal- and gene-therapeutic agents via different mechanisms. Herbal ingredients can affect different signaling pathways, reduce the toxic side effects or inhibit the efflux of anticancer drugs.Areas covered: This review will discuss the delivery of herbal compounds with other cancer treatments such as hormonal, small molecule inhibitors and inorganic hybrids to tumor cells. An overview of physicochemical properties of herbal components that require intelligent design of combo-nanomedicines for efficient co-delivery of those herbal-derived and other anticancer agents was discussed. Nanocarriers provide various benefits to overcome the shortcomings of the encapsulated herbal compounds including improved solubility, increased stability and enhanced tumor targeting. Different nanocarrier systems were the focus of this review.Expert opinion: Multifunctional nanocarrier systems encapsulating herbal and different anticancer drugs showed to be a wonderful approach in the treatment of cancer enabling the co-delivery of anticancer drugs with versatile modes of action in an accurate manner in an attempt to enhance the efficacy, benefit from the synergism between the drugs as well as to minimize the development of multi-drug resistance. The main challenge point is the early detection and management of any developed adverse effect.
Collapse
Affiliation(s)
- Doaa M Anwar
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Arab Academy for Science Technology & Maritime Transport, Alexandria, Egypt.,Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mousa El-Sayed
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt.,Department of Chemistry, School of Sciences and Engineering, American University in Cairo, New Cairo, Egypt
| | - Asmaa Reda
- Nanomedicine Division, Center for Materials Science, University of Science and Technology (UST), Zewail City of Science and Technology, Giza, Egypt.,Molecular and Cellular Biology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Research Center for Industry of Human Ecology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of AnesthesiologyChang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Sherine N Khattab
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.,Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
49
|
Yin F, Xu F, Zhang K, Yuan M, Cao H, Ye T, Wu X, Xu F. Synthesis and evaluation of mesoporous silica/mesoporous molecularly imprinted nanoparticles as adsorbents for detection and selective removal of imidacloprid in food samples. Food Chem 2021; 364:130216. [PMID: 34237619 DOI: 10.1016/j.foodchem.2021.130216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/07/2021] [Accepted: 05/17/2021] [Indexed: 11/18/2022]
Abstract
The double-mesoporous-layer imprinted polymer of mesoporous silica/mesoporous molecularly imprinted nanoparticles (MIP), with high specific surface area, rich porosity, excellent mass transfer rate and selectivity, were synthesized using imidacloprid (IDP) as a template. Under the optimal conditions of pH, contact time, concentration and temperature, MIP showed high adsorption capacity of 13.86 μg·mg-1 toward IDP and the imprinting factor reached 3.5. The adsorption process model including binding isotherm and kinetics was investigated. MIP exhibited excellent regeneration and its adsorption and selectivity were outstanding among its structurally pesticide analogues. The recovery of spiked IDP for MIP in fortified real samples can reach 96.0 ± 8.5% for cabbage and 105.0 ± 9.9% for apple. The limit of detection of the enrichment method can be as low as 0.037 μg·mL-1 with a good linear relationship (R2 = 0.996) from 0.30 to 10.0 μg·mL-1. The results indicated that the proposed method allowed class-specific detection of IDP in food samples.
Collapse
Affiliation(s)
- Fengqin Yin
- School of Medical Instrument and Food Engineering, Shanghai Engineering Research Center for Food Rapid Detection, University of Shanghai for Science and Technology, Shanghai, China
| | - Feng Xu
- College of Science, University of Shanghai for Science and Technology, Shanghai, China
| | - Kun Zhang
- College of Science, University of Shanghai for Science and Technology, Shanghai, China
| | - Min Yuan
- School of Medical Instrument and Food Engineering, Shanghai Engineering Research Center for Food Rapid Detection, University of Shanghai for Science and Technology, Shanghai, China
| | - Hui Cao
- School of Medical Instrument and Food Engineering, Shanghai Engineering Research Center for Food Rapid Detection, University of Shanghai for Science and Technology, Shanghai, China
| | - Tai Ye
- School of Medical Instrument and Food Engineering, Shanghai Engineering Research Center for Food Rapid Detection, University of Shanghai for Science and Technology, Shanghai, China
| | - Xiuxiu Wu
- School of Medical Instrument and Food Engineering, Shanghai Engineering Research Center for Food Rapid Detection, University of Shanghai for Science and Technology, Shanghai, China
| | - Fei Xu
- School of Medical Instrument and Food Engineering, Shanghai Engineering Research Center for Food Rapid Detection, University of Shanghai for Science and Technology, Shanghai, China; College of Science, University of Shanghai for Science and Technology, Shanghai, China.
| |
Collapse
|
50
|
Asrorov AM, Gu Z, Li F, Liu L, Huang Y. Biomimetic camouflage delivery strategies for cancer therapy. NANOSCALE 2021; 13:8693-8706. [PMID: 33949576 DOI: 10.1039/d1nr01127h] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer remains a significant challenge despite the progress in developing different therapeutic approaches. Nanomedicine has been explored as a promising novel cancer therapy. Recently, biomimetic camouflage strategies have been investigated to change the bio-fate of therapeutics and target cancer cells while reducing the unwanted exposure on normal tissues. Endogenous components (e.g., proteins, polysaccharides, and cell membranes) have been used to develop anticancer drug delivery systems. These biomimetic systems can overcome biological barriers and enhance tumor cell-specific uptake. The tumor-targeting mechanisms include ligand-receptor interactions and stimuli-responsive (e.g., pH-sensitive and light-sensitive) delivery. Drug delivery carriers composed of endogenous components represent a promising approach for improving cancer treatment efficacy. In this paper, different biomimetic drug delivery strategies for cancer treatment are reviewed with a focus on the discussion of their advantages and potential applications.
Collapse
Affiliation(s)
- Akmal M Asrorov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China. and Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, 83, M. Ulughbek Street, Tashkent 100125, Uzbekistan
| | - Zeyun Gu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | - Feng Li
- Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA.
| | - Lingyun Liu
- First Clinical School, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China. and Zhongshan Institute for Drug Discovery, Institutes of Drug Discovery and Development, Chinese Academy of Sciences, Zhongshan 528437, China and NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China
| |
Collapse
|