51
|
Abd-Rabou AA, Kishta MS, Yakout SM, Youssef AM, Abdallah AN, Ahmed HH. Copper/Tin Nanocomposites-Loaded Exosomes Induce Apoptosis and Cell Cycle Arrest at G0/G1 Phase in Skin Cancer Cell Line. Chem Biodivers 2024; 21:e202400486. [PMID: 38860853 DOI: 10.1002/cbdv.202400486] [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: 02/29/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/12/2024]
Abstract
This study aims to explore the efficacy of Copper/Tin (CuS/SnS) nanocomposites loaded into exosomes against skin cancer A431 cell line. CuS/SnS nanocomposites (S1, S2, S3) were synthesized and characterized, then loaded into exosomes (Exo) (S1-Exo, S2-Exo and S3-Exo) and characterized. After that, the loaded samples were investigated in vitro against A431 using cytotoxicity, apoptosis, and cell cycle assays. CuS/SnS nanocomposites were indexed to hexagonal CuS structure and orthorhombic α-SnS phase and showed nano-rode shape. The exosomes loaded with nanocomposites were regular and rounded within the size of 120 nm, with no signs of broken exosomes or leakage of their contents. The cytotoxicity assay indicated the enhanced cytotoxic of S1-Exo versus the free nano-form S1 on A431. Interestingly, S1-Exo recorded 1.109 times more than DOX in its anti-skin cancer capacity. Moreover, S1-Exo recorded 40.2 % for early apoptosis and 22.1 % for late apoptosis. Furthermore, it displayed impact in arresting the cancer cell cycle at G0/G1 phase and reducing G2/M phase. Noteworthy, loaded nanocomposites were safe against normal HSF skin cells. In conclusion, the loaded CuS/SnS nanocomposites into the exosomes could be of great potential as anti-skin cancer candidates through induction of apoptosis and promotion of the cell cycle arrest at G0/G1 phase.
Collapse
Affiliation(s)
- Ahmed A Abd-Rabou
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, Egypt
- Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Mohamed S Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, Egypt
- Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Saad M Yakout
- Inorganic Chemistry Department, Inorganic Chemical Industries and Mineral Resources Research institute, National Research Centre, Dokki, Cairo, Egypt
| | - Ahmed M Youssef
- Inorganic Chemistry Department, Inorganic Chemical Industries and Mineral Resources Research institute, National Research Centre, Dokki, Cairo, Egypt
| | - Ahmed N Abdallah
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, Egypt
- Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Hanaa H Ahmed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, Egypt
- Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| |
Collapse
|
52
|
Liao J, Timoshenko AB, Cordova DJ, Astudillo Potes MD, Gaihre B, Liu X, Elder BD, Lu L, Tilton M. Propelling Minimally Invasive Tissue Regeneration With Next-Era Injectable Pre-Formed Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400700. [PMID: 38842622 DOI: 10.1002/adma.202400700] [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: 01/14/2024] [Revised: 05/12/2024] [Indexed: 06/07/2024]
Abstract
The growing aging population, with its associated chronic diseases, underscores the urgency for effective tissue regeneration strategies. Biomaterials play a pivotal role in the realm of tissue reconstruction and regeneration, with a distinct shift toward minimally invasive (MI) treatments. This transition, fueled by engineered biomaterials, steers away from invasive surgical procedures to embrace approaches offering reduced trauma, accelerated recovery, and cost-effectiveness. In the realm of MI tissue repair and cargo delivery, various techniques are explored. While in situ polymerization is prominent, it is not without its challenges. This narrative review explores diverse biomaterials, fabrication methods, and biofunctionalization for injectable pre-formed scaffolds, focusing on their unique advantages. The injectable pre-formed scaffolds, exhibiting compressibility, controlled injection, and maintained mechanical integrity, emerge as promising alternative solutions to in situ polymerization challenges. The conclusion of this review emphasizes the importance of interdisciplinary design facilitated by synergizing fields of materials science, advanced 3D biomanufacturing, mechanobiological studies, and innovative approaches for effective MI tissue regeneration.
Collapse
Affiliation(s)
- Junhan Liao
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Anastasia B Timoshenko
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Domenic J Cordova
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Benjamin D Elder
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maryam Tilton
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
53
|
Gao P, Duan Z, Xu G, Gong Q, Wang J, Luo K, Chen J. Harnessing and Mimicking Bacterial Features to Combat Cancer: From Living Entities to Artificial Mimicking Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405075. [PMID: 39136067 DOI: 10.1002/adma.202405075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/23/2024] [Indexed: 08/29/2024]
Abstract
Bacterial-derived micro-/nanomedicine has garnered considerable attention in anticancer therapy, owing to the unique natural features of bacteria, including specific targeting ability, immunogenic benefits, physicochemical modifiability, and biotechnological editability. Besides, bacterial components have also been explored as promising drug delivery vehicles. Harnessing these bacterial features, cutting-edge physicochemical and biotechnologies have been applied to attenuated tumor-targeting bacteria with unique properties or functions for potent and effective cancer treatment, including strategies of gene-editing and genetic circuits. Further, the advent of bacteria-inspired micro-/nanorobots and mimicking artificial systems has furnished fresh perspectives for formulating strategies for developing highly efficient drug delivery systems. Focusing on the unique natural features and advantages of bacteria, this review delves into advances in bacteria-derived drug delivery systems for anticancer treatment in recent years, which has experienced a process from living entities to artificial mimicking systems. Meanwhile, a summary of relative clinical trials is provided and primary challenges impeding their clinical application are discussed. Furthermore, future directions are suggested for bacteria-derived systems to combat cancer.
Collapse
Affiliation(s)
- Peng Gao
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Gang Xu
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, 361000, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kui Luo
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Jie Chen
- Department of General Surgery, Breast Disease Center, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
54
|
Gao F, Guo J, Liu S, Zhang F, Zhang Y, Wang L. Empowering hydrophobic anticancer drugs by ultrashort peptides: General Co-assembly strategy for improved solubility, targeted efficacy, and clinical application. J Colloid Interface Sci 2024; 667:119-127. [PMID: 38631250 DOI: 10.1016/j.jcis.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
The current state of drug delivery systems allows for the resolution of specific issues like inadequate solubility, limited targeting capabilities, and complex preparation processes, requiring tailored designs for different drugs. Yet, the major challenge in clinical application lies in surmounting these obstacles with a universal carrier that is effective for a variety of anticancer drugs. Herein, with the help of computer simulation, we rationally design ultrashort peptides GY and CCYRGD, which can co-assemble with hydrophobic anticancer drugs into nanoparticles with enhanced solubility, targeting ability and anticancer efficacy. Taking 7-ethyl-10-hydroxy camptothecin (SN38) as a model anticancer drug, the co-assembled SN38-GY-CCYRGD nanoparticles significantly enhance the water solubility of SN38 by more than three orders of magnitude. The as-prepared nanoparticles can effectively kill cancer cells, e.g., human small cell lung cancer (A549) cells with a notable cell mortality rate of 71%. Mice experimental results demonstrate the nanoparticles' efficient targeting capability, marked reducing the toxicity to normal tissues while improving antitumor efficacy. This work presents a novel drug delivery method, integrating effective, targeted, and safe strategies into a comprehensive carrier system, designed for the administration of hydrophobic anticancer drugs.
Collapse
Affiliation(s)
- Feng Gao
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China; Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Jun Guo
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Shihao Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Feng Zhang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China; Key Laboratory of Optical Technology and Instrument for Medicine, Ministry of Education, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yi Zhang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
| | - Liping Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China.
| |
Collapse
|
55
|
Nešić MD, Dučić T, Gemović B, Senćanski M, Algarra M, Gonçalves M, Stepić M, Popović IA, Kapuran Đ, Petković M. Prediction of Protein Targets in Ovarian Cancer Using a Ru-Complex and Carbon Dot Drug Delivery Therapeutic Nanosystems: A Bioinformatics and µ-FTIR Spectroscopy Approach. Pharmaceutics 2024; 16:997. [PMID: 39204341 PMCID: PMC11359177 DOI: 10.3390/pharmaceutics16080997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
We predicted the protein therapeutic targets specific to a Ru-based potential drug and its combination with pristine and N-doped carbon dot drug delivery systems, denoted as RuCN/CDs and RuCN/N-CDs. Synchrotron-based FTIR microspectroscopy (µFTIR) in addition to bioinformatics data on drug structures and protein sequences were applied to assess changes in the protein secondary structure of A2780 cancer cells. µFTIR revealed the moieties of the target proteins' secondary structure changes only after the treatment with RuCN and RuCN/N-CDs. A higher content of α-helices and a lower content of β-sheets appeared in A2780 cells after RuCN treatment. Treatment with RuCN/N-CDs caused a substantial increase in parallel β-sheet numbers, random coil content, and tyrosine residue numbers. The results obtained suggest that the mitochondrion-related proteins NDUFA1 and NDUFB5 are affected by RuCN either via overexpression or stabilisation of helical structures. RuCN/N-CDs either induce overexpression of the β-sheet-rich protein NDUFS1 and affect its random coil structure or interact and stabilise its structure via hydrogen bonding between -NH2 groups from N-CDs with protein C=O groups and -OH groups of serine, threonine, and tyrosine residues. The N-CD nanocarrier tunes this drug's action by directing it toward a specific protein target, changing this drug's coordination ability and inducing changes in the protein's secondary structures and function.
Collapse
Affiliation(s)
- Maja D. Nešić
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (I.A.P.); (Đ.K.); (M.P.)
| | - Tanja Dučić
- ALBA-CELLS Synchrotron, 08290 Cerdanyola del Vallès, Spain
| | - Branislava Gemović
- Laboratory for Bioinformatics and Computational Chemistry, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.G.); (M.S.)
| | - Milan Senćanski
- Laboratory for Bioinformatics and Computational Chemistry, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.G.); (M.S.)
| | - Manuel Algarra
- INAMAT2—Institute for Advanced Materials and Mathematics, Department of Science, Public University of Navarre, Campus de Arrosadia, 31006 Pamplona, Spain;
| | - Mara Gonçalves
- CQM—Centro de Química da Madeira, Universidade da Madeira, 9020-105 Funchal, Portugal;
| | - Milutin Stepić
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (I.A.P.); (Đ.K.); (M.P.)
| | - Iva A. Popović
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (I.A.P.); (Đ.K.); (M.P.)
| | - Đorđe Kapuran
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (I.A.P.); (Đ.K.); (M.P.)
| | - Marijana Petković
- Center for Light-Based Research and Technologies COHERENCE, Department of Atomic Physics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (I.A.P.); (Đ.K.); (M.P.)
| |
Collapse
|
56
|
Bhuker S, Kaur A, Rajauria K, Tuli HS, Saini AK, Saini RV, Gupta M. Allicin: a promising modulator of apoptosis and survival signaling in cancer. Med Oncol 2024; 41:210. [PMID: 39060753 DOI: 10.1007/s12032-024-02459-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
According to the World Health Organization, cancer is the foremost cause of mortality globally. Various phytochemicals from natural sources have been extensively studied for their anticancer properties. Allicin, a powerful organosulfur compound derived from garlic, exhibits anticancer, antioxidant, anti-inflammatory, antifungal, and antibacterial properties. This review aims to update and evaluate the chemistry, composition, mechanisms of action, and pharmacokinetics Allicin. Allicin has garnered significant attention for its potential role in modulating Fas-FasL, Bcl2-Bax, PI3K-Akt-mTOR, autophagy, and miRNA pathways. At the molecular level, allicin induces the release of cytochrome c from the mitochondria and enhances the activation of caspases-3, -8, and -9. This is accompanied by the simultaneous upregulation of Bax and Fas expression in tumor cells. Allicin can inhibit excessive autophagy by activating the PI3K/Akt/mTOR and MAPK/ERK/mTOR signaling pathways. Allicin-loaded nano-formulations efficiently induce apoptosis in cancer cells while minimizing toxicity to normal cells. Safety and clinical aspects are meticulously scrutinized, providing insights into the tolerability and adverse effects associated with allicin administration, along with an overview of current clinical trials evaluating its therapeutic potential. In conclusion, this review underscores the promising prospects of allicin as a dietary-derived medicinal compound for cancer therapy. It emphasizes the need for further research to elucidate its precise mechanisms of action, optimize delivery strategies, and validate its efficacy in clinical settings.
Collapse
Affiliation(s)
- Sunaina Bhuker
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
| | - Avneet Kaur
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
| | - Kanitha Rajauria
- SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Tamil Nadu, 603203, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
| | - Adesh K Saini
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
- Central Research Laboratory, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India
| | - Reena V Saini
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India.
- Central Research Laboratory, Maharishi Markandeshwar (Deemed to Be University), Mullana, Haryana, 133207, India.
- Central Research Laboratory and Department of Bio-Sciences and Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana, 133207, India.
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India
| |
Collapse
|
57
|
Singh S, Tiwari H, Verma A, Gupta P, Chattopadhaya A, Singh A, Singh S, Kumar B, Mandal A, Kumar R, Yadav AK, Gautam HK, Gautam V. Sustainable Synthesis of Novel Green-Based Nanoparticles for Therapeutic Interventions and Environmental Remediation. ACS Synth Biol 2024; 13:1994-2007. [PMID: 38899943 DOI: 10.1021/acssynbio.4c00206] [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] [Indexed: 06/21/2024]
Abstract
The advancement in nanotechnology has completely revolutionized various fields, including pharmaceutical sciences, and streamlined the potential therapeutic of many diseases that endanger human life. The synthesis of green nanoparticles by biological processes is an aspect of the newly emerging scientific field known as "green nanotechnology". Due to their safe, eco-friendly, nontoxic nature, green synthesis tools are better suited to produce nanoparticles between 1 and 100 nm. Nanoformulation of different types of nanoparticles has been made possible by using green production techniques and commercially feasible novel precursors, such as seed extracts, algae, and fungi, that act as potent reducing, capping, and stabilizing agents. In addition to this, the biofunctionalization of nanoparticles has also broadened its horizon in the field of environmental remediation and various novel therapeutic innovations including wound healing, antimicrobial, anticancer, and nano biosensing. However, the major challenge pertaining to green nanotechnology is the agglomeration of nanoparticles that may alter the surface topology, which can affect biological physiology, thereby contributing to system toxicity. Therefore, a thorough grasp of nanoparticle toxicity and biocompatibility is required to harness the applications of nanotechnology in therapeutics.
Collapse
Affiliation(s)
- Swati Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Harshita Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Ashish Verma
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Priyamvada Gupta
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Amrit Chattopadhaya
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Ananya Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
- Department of Botany, Faculty of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Sanjana Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
- Department of Botany, Faculty of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Brijesh Kumar
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, 221005, India
| | - Abhijit Mandal
- Department of Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Ashok K Yadav
- Centre for Molecular Biology, Central University of Jammu, Samba, 181143, Jammu and Kashmir (UT), India
| | - Hemant Kumar Gautam
- Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, New Delhi 110025, India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
58
|
Varpe P, Joga R, Aglave G, Vasu P, Yerram S, Bellapu KK, Srivastava S, Kumar S. Esterase responsive release of anti-cancer agents from conjugated lipid nanocarrier and the regulatory considerations. Pharm Pat Anal 2024; 13:31-43. [PMID: 39324857 PMCID: PMC11449025 DOI: 10.1080/20468954.2024.2347796] [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: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 09/27/2024]
Abstract
The release of active agents in tumors rather than normal tissues, limits systemic exposure and toxicities. Targeting over-expressed esterase enzyme in the tumor microenvironment can selectively release immune-active agents like Programmed Death-1 (PD-1) and PD-1 ligand inhibitors from ester-sensitive lipid nanocarriers, offering a novel approach compared with conventional therapies. PD-1 and PD-L1 association cause T-cell inactivation, whereas blocking their association improves their cytotoxic mechanism. The patent application US2022/0080051-A1 discloses a novel immune-active agent conjugated with lipid to form a nanocarrier for esterase-sensitive release. These nanocarriers selectively enter leaky vasculature of tumors through enhanced permeability and retention effect, undergo ester cleavage to release agents, and are reported to increase bioavailability by 24 times. Further, with other agents or alone it achieves targeted synergistic cancer therapy. Also, the current patent spotlight delves into the crucial formulation considerations necessary for obtaining successful approval of lipidic nano products from relevant regulatory authorities.
Collapse
Affiliation(s)
- Priya Varpe
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Gayatri Aglave
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Pavan Vasu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Sravani Yerram
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Kiran Kumar Bellapu
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana , 500037, India
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar, Hyderabad, Telangana, 500037, India
- Department of Pharmaceutics, Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur, Rajasthan 303121, India
| |
Collapse
|
59
|
Alhakamy NA, Abdullah S, Md S, Ansari AR, Bhattamisra SK, Ibrahim IM, Alahdal H, Altamimi AA, Shaik RA. Oral co-polymeric raft-forming nano gels for targeted empagliflozin delivery against stomach cancer (SGC7901). Heliyon 2024; 10:e34074. [PMID: 39071709 PMCID: PMC11279758 DOI: 10.1016/j.heliyon.2024.e34074] [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: 08/06/2023] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
Empagliflozin (EMP) is known for its poor safety and efficacy profile due to its fast body distribution and poor solubility. Accordingly, an oral long-acting and floating/raft-forming nano gel was optimized to release coated EMP nanoparticles, and the released EMP nanoparticles showed enhanced dissolution compared to raw EMP particles. To repurpose EMP for cancer treatment, EMP shows anti-cancer and anti-inflammatory effects against cancer cells. EMP nanoparticles were characterized using FT-IR, PXRD, SEM, EMP encapsulation assay, and release studies. The raft-forming gel encapsulating the EMP was optimized and characterized. The EMP co-polymeric nanoparticles were studied to investigate EMP anti-cancer and anti-inflammatory activities against stomach cancer cells. The solubility of EMP nanoparticles was enhanced in 0.1 N HCl and pH 6.8 by 5 and 12 folds, respectively, compared to raw EMP powder. The particle size and zeta-potential values of improved EMP nanoparticles were 135.40 ± 18.60 nm, and -19.30 ± 0.80 mV, respectively. FT-IR, PXRD, SEM and TEM characterizations revealed polymeric coating of EMP particles. The study suggested that this optimized controlled-release raft-forming gel is a promising local oral approach against stomach cancer. The repurposing of EMP co-polymeric nanoparticles for stomach cancer and associated gastritis treatment was justified.
Collapse
Affiliation(s)
- Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Center of Excellence for Drug Research Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samaa Abdullah
- Natural and Health Sciences Research Centre, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Center of Excellence for Drug Research Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Subrat Kumar Bhattamisra
- Department of Pharmaceutical Technology, School of Medical Science, Adamas University, Kolkata, India
| | - Ibrahim M. Ibrahim
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hadil Alahdal
- Department of Biology, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, 84428, Saudi Arabia
| | - Abeer A. Altamimi
- Natural and Health Sciences Research Centre, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Rasheed A. Shaik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
60
|
Kong JC, Zhou F, Shi L, Wei Y, Wu C. A novel nanodrug for the sensitization of photothermal chemotherapy for breast cancer in vitro. RSC Adv 2024; 14:21292-21299. [PMID: 38974230 PMCID: PMC11225340 DOI: 10.1039/d4ra01611d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024] Open
Abstract
Owing to the complexity of tumor treatment, clinical tumor treatment has evolved from a single treatment mode to multiple combined treatment modes. Reducing the tolerance of tumors to heat and the toxicity of chemotherapy drugs to the body, as well as increasing the sensitivity of tumors to photothermal therapy and chemotherapy drugs, are key issues that urgently need to be addressed in the current cancer treatment. In this work, polylactic acid-based drug nanoparticles (PLA@DOX/GA/ICG) were synthesized with good photothermal conversion ability by encapsulating the water-soluble anticancer drug doxorubicin (DOX), photothermal conversion agent indocyanine green (ICG) and liposoluble drug gambogic acid (GA) using a double emulsion method. The preparation process of PLA@DOX/GA/ICG was examined. Gambogic acid entrapped in PLA@DOX/GA/ICG nanoparticles could act as an HSP90 protein inhibitor to achieve bidirectional sensitization to chemotherapy and photothermal therapy under 808 nm laser irradiation for the first time, effectively ablating breast cancer cells in vitro. This nanodrug was expected to be used for the efficient treatment of tumors.
Collapse
Affiliation(s)
- Ji Chuan Kong
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Feng Zhou
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Liting Shi
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Yihui Wei
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Chunhong Wu
- Henan Polytechinc University Jiaozuo Henan 45400 China
| |
Collapse
|
61
|
Oehler JB, Rajapaksha W, Albrecht H. Emerging Applications of Nanoparticles in the Diagnosis and Treatment of Breast Cancer. J Pers Med 2024; 14:723. [PMID: 39063977 PMCID: PMC11278299 DOI: 10.3390/jpm14070723] [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: 05/18/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer remains the most prevalent cancer among women worldwide, driving the urgent need for innovative approaches to diagnosis and treatment. This review highlights the pivotal role of nanoparticles in revolutionizing breast cancer management through advancements of interconnected approaches including targeted therapy, imaging, and personalized medicine. Nanoparticles, with their unique physicochemical properties, have shown significant promise in addressing current treatment limitations such as drug resistance and nonspecific systemic distribution. Applications range from enhancing drug delivery systems for targeted and sustained release to developing innovative diagnostic tools for early and precise detection of metastases. Moreover, the integration of nanoparticles into photothermal therapy and their synergistic use with existing treatments, such as immunotherapy, illustrate their transformative potential in cancer care. However, the journey towards clinical adoption is fraught with challenges, including the chemical feasibility, biodistribution, efficacy, safety concerns, scalability, and regulatory hurdles. This review delves into the current state of nanoparticle research, their applications in breast cancer therapy and diagnosis, and the obstacles that must be overcome for clinical integration.
Collapse
Affiliation(s)
- Josephine B. Oehler
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4810, Australia
- Biomedical Sciences and Molecular Biology, College of Public Health, Medical & Vet Sciences, James Cook University, Townsville, QLD 4810, Australia
| | - Weranga Rajapaksha
- Centre for Pharmaceutical Innovation (CPI), Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Hugo Albrecht
- Centre for Pharmaceutical Innovation (CPI), Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| |
Collapse
|
62
|
Andrés CMC, Pérez de la Lastra JM, Bustamante Munguira E, Andrés Juan C, Pérez-Lebeña E. Anticancer Activity of Metallodrugs and Metallizing Host Defense Peptides-Current Developments in Structure-Activity Relationship. Int J Mol Sci 2024; 25:7314. [PMID: 39000421 PMCID: PMC11242492 DOI: 10.3390/ijms25137314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
This article provides an overview of the development, structure and activity of various metal complexes with anti-cancer activity. Chemical researchers continue to work on the development and synthesis of new molecules that could act as anti-tumor drugs to achieve more favorable therapies. It is therefore important to have information about the various chemotherapeutic substances and their mode of action. This review focuses on metallodrugs that contain a metal as a key structural fragment, with cisplatin paving the way for their chemotherapeutic application. The text also looks at ruthenium complexes, including the therapeutic applications of phosphorescent ruthenium(II) complexes, emphasizing their dual role in therapy and diagnostics. In addition, the antitumor activities of titanium and gold derivatives, their side effects, and ongoing research to improve their efficacy and reduce adverse effects are discussed. Metallization of host defense peptides (HDPs) with various metal ions is also highlighted as a strategy that significantly enhances their anticancer activity by broadening their mechanisms of action.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | | | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain
| | | |
Collapse
|
63
|
Kiruthiga C, Balan DJ, Prasath NH, Manikandakrishnan M, Jafni S, Prabhu NM, Pandian SK, Devi KP. Synergistic induction of apoptosis in lung cancer cells through co-delivery of PLGA phytol/α-bisabolol nanoparticles. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5131-5144. [PMID: 38240783 DOI: 10.1007/s00210-023-02935-2] [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: 08/02/2023] [Accepted: 12/27/2023] [Indexed: 06/12/2024]
Abstract
This study explored the potential of poly-(lactic-co-glycolic) acid (PLGA) nanoparticles to enhance the effectiveness of anticancer treatments through combination therapy with phytol and α-bisabolol. The encapsulation efficiency of the nanoparticles was investigated, highlighting the role of ionic interactions between the drugs and the polymer. Characterization of PLGA-Phy+Bis nanoparticles was carried out using DLS with zeta potential and HR-TEM for size determination. Spectrophotometric measurements evaluated the encapsulation efficiency, loading efficiency, and in vitro drug release. FTIR analysis assessed the chemical interactions between PLGA and the drug actives, ensuring nanoparticle stability. GC-MS was employed to analyze the chemical composition of drug-loaded PLGA nanocarriers. Cytotoxicity was evaluated via the MTT assay, while Annexin V-FITC/PI staining and western blot analysis confirmed apoptotic cell death. Additionally, toxicity tests were performed on L-132 cells and in vivo zebrafish embryos. The study demonstrates high encapsulation efficiency of PLGA-Phy+Bis nanoparticles, which exhibit monodispersity and sizes of 189.3±5nm (DLS) and 268±54 nm (HR-TEM). Spectrophotometric analysis confirmed efficient drug encapsulation and release control. FTIR analysis revealed nanoparticle structural stability without chemical interactions. MTT assay results demonstrated the promising anticancer potential of all the three nanoparticle types (PLGA-Phy, PLGA-Bis, and PLGA-Phy+Bis) against lung cancer cells. Apoptosis was confirmed through Annexin V-FITC/PI staining and western blot analysis, which also revealed changes in Bax and Bcl-2 protein expression. Furthermore, the nanoparticles exhibited non-toxicity in L-132 cells and zebrafish embryo toxicity tests. PLGA-Phy+Bis nanoparticles exhibited efficient encapsulation, controlled release, and low toxicity. Apoptosis induction in A549 cells and non-toxicity in healthy cells highlight their clinical potential.
Collapse
Affiliation(s)
| | | | - Nagaiah Hari Prasath
- Department of Biotechnology, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India
| | - Muthushanmugam Manikandakrishnan
- Disease Control and Prevention lab, Department of Animal Health and Management, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India
| | - Sakthivel Jafni
- Department of Biotechnology, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India
| | - Narayanasamy Marimuthu Prabhu
- Disease Control and Prevention lab, Department of Animal Health and Management, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India
| | | | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India.
| |
Collapse
|
64
|
Mahdavi Niyaki Z, Salehzadeh A, Peymani M, Zaefizadeh M. Exploring the Therapeutic Potential of Fe 3O 4@Glu-Oleuropein Nanoparticles in Targeting KRAS Pathway-Regulating lncRNAs in Colorectal Cancer Cells. Biol Trace Elem Res 2024; 202:3073-3085. [PMID: 37792268 DOI: 10.1007/s12011-023-03892-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
Cancer, the leading cause of death worldwide, has witnessed significant advancements in treatment through targeted therapies. Among the proto-oncogenes prevalent in human cancers, KRAS stands out, and recent research has focused on long noncoding RNAs (lncRNAs) as regulators of miRNAs targeting the KRAS oncogene. This study specifically explores lncRNAs involved in the KRAS pathway in colorectal cancer (CRC). To investigate this, researchers employed iron oxide nanoparticles coated with glucose and conjugated with Oleuropein (Fe3O4@Glu-Oleuropein NPs) to evaluate their impact on candidate lncRNAs associated with KRAS pathway deregulation. The study utilized TCGA data to identify genes affected by KRAS mutation and lncRNAs linked to KRAS in CRC. Enrichr and MsigDB databases helped identify relevant pathways. Genes with a correlation coefficient above 0.5 and a P-value less than 0.01 with candidate lncRNAs were selected. MTT and flow cytometry assays determined the anti-proliferative and apoptotic effects of Fe3O4@Glu-Oleuropein NPs on CRC cells (SW480) and normal cells (HEK293). The findings showed that increased expression of FEZF1-AS1, GAS6-AS1, and LINC00920 correlated with mutated KRAS, and co-expressed genes were significantly involved in hypoxia, KRAS signaling, DNA repair, and IL-2/STAT5 signaling pathways. Fe3O4@Glu-Oleuropein NPs exhibited higher toxicity toward cancer cells, with IC50 values of 92 μg/ml for SW480 and 281 μg/ml for HEK293. Flow cytometry analysis revealed a substantial increase in necrotic and apoptotic cells when treated with Fe3O4@Glu-Oleuropein, along with down-regulation of GAS6-AS1, LINC00920, and FEZF1-AS1 lncRNAs in treated cells. In conclusion, this study highlights the therapeutic potential of Fe3O4@Glu-Oleuropein on colon cancer cells in vitro. The identification of lncRNAs involved in the KRAS pathway provides insights into the underlying mechanisms and offers avenues for further research in targeted cancer therapies.
Collapse
Affiliation(s)
| | - Ali Salehzadeh
- Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran.
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mohammad Zaefizadeh
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| |
Collapse
|
65
|
Alali U, Al-Tu’ma MMK, Jawad AF, Ahmed ST, Ali HA, Abdulzehra S, Al-Tu’ma FJ. Synthesis, Characterization, and In-Vitro Evaluation of Silibinin-loaded PEGylated Niosomal Nanoparticles: Potential Anti-Cancer Effects on SW480 Colon Cancer Cells. Asian Pac J Cancer Prev 2024; 25:2539-2550. [PMID: 39068589 PMCID: PMC11480629 DOI: 10.31557/apjcp.2024.25.7.2539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE Colorectal cancer is a significant global health concern with high mortality rates. Silibinin is a compound derived from milk thistle with anticancer properties and may be a potential treatment option for colorectal cancer. Its poor solubility limits its clinical application, but various strategies, such as nanoparticle encapsulation, have shown promise. In this study, a PEGylated niosomal drug delivery system was used to enhance the solubility of silibinin, and its anti-proliferative effects were evaluated against human colorectal cancer cell lines. METHODS The silibinin-loaded PEGylated niosomal nanoparticles (NIO-SIL) were fabricated using the thin-film hydration method and characterized with dialysis bag, AFM, SEM, DLS, and FTIR systems. Finally, the cancerous cells and human normal cells were treated with NIO-SIL and pure silibinin. The proliferation, apoptosis, and cell cycle of these cells were evaluated. Subsequently, the expression of Bax, Bcl-2, p53, and cyclin D1 genes was measured using real-time PCR. RESULT The drug release profile, size, morphology, and chemical interactions of the synthesized PEGylated niosomal nanoparticles were suitable for use as a drug delivery system. Both pure silibinin and NIO-SIL could reduce the proliferation of cancerous cells, induce apoptosis, and cause cell cycle arrest, with no significant negative effects reported on human normal cells. Both pure silibinin and NIO-SIL reduced the expression of the Bcl-2 and cyclin D1 genes while increasing the expression of Bax and p53. (p-value < 0.05 *). CONCLUSION The outcomes of this study indicate the high potential of PEGylated niosomal nanoparticles for encapsulation and delivery of silibinin to cancer cells, with no negative effects on normal cells.
Collapse
Affiliation(s)
- Urjwan Alali
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Kerbala, Kerbala, Iraq.
| | - Maha Mohammed Kadhim Al-Tu’ma
- Department of Anesthesia Techniques, College of Health and Medical Techniques, Al-Zahraa University for Women, Kerbala, Iraq.
| | - Ammar Fadhil Jawad
- Department of Pharmacognesy, College of Pharmacy, University of Kerbala, Kerbala, Iraq.
| | - Saja Talib Ahmed
- Department of Chemistry, College of Science, University of Kufa, Kufa, Iraq.
| | - Hanaa Addai Ali
- Department of Chemistry, College of Science, University of Kufa, Kufa, Iraq.
| | - Siham Abdulzehra
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fadhil Jawad Al-Tu’ma
- Department of Chemistry and Biochemistry, College of Medicine, University of Kerbala, Kerbala, Iraq.
| |
Collapse
|
66
|
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
|
67
|
Waqar MA, Zaman M, Khan R, Shafeeq Ur Rahman M, Majeed I. Navigating the tumor microenvironment: mesenchymal stem cell-mediated delivery of anticancer agents. J Drug Target 2024; 32:624-634. [PMID: 38652480 DOI: 10.1080/1061186x.2024.2347356] [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: 02/26/2024] [Accepted: 04/21/2024] [Indexed: 04/25/2024]
Abstract
Scientific knowledge of cancer has advanced greatly throughout the years, with most recent studies findings includes many hallmarks that capture disease's multifaceted character. One of the novel approach utilised for the delivery of anti-cancer agents includes mesenchymal stem cell mediated drug delivery. Mesenchymal stem cells (MSCs) are non-haematopoietic progenitor cells that may be extracted from bone marrow, tooth pulp, adipose tissue and placenta/umbilical cord blood dealing with adult stem cells. MSCs are mostly involved in regeneration of tissue, they have also been shown to preferentially migrate to location of several types of tumour in-vivo. Usage of MSCs ought to improve both effectiveness and safety of anti-cancer drugs by enhancing delivery efficiency of anti-cancer therapies to tumour site. Numerous researches has demonstrated that various drugs, when delivered via mesenchymal stem cell mediated delivery can elicit anti-tumour effect of cells in cancers of breast cells and thyroid cells. MSCs have minimal immunogenicity because to lack of co-stimulatory molecule expression, which means there is no requirement for immunosuppression after allogenic transplantation. This current review elaborates recent advancements of mesenchyma stem cell mediated drug delivery of anti-cancer agents along with its mechanism and previously reported studies of drugs manufactured via this drug delivery system.
Collapse
Affiliation(s)
- Muhammad Ahsan Waqar
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | - Muhammad Zaman
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Rabeel Khan
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | | | - Imtiaz Majeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
68
|
Natallia L, Dama A, Gorica E, Darya K, Peña-Corona SI, Cortés H, Santini A, Büsselberg D, Leyva-Gómez G, Sharifi-Rad J. Genipin's potential as an anti-cancer agent: from phytochemical origins to clinical prospects. Med Oncol 2024; 41:186. [PMID: 38918260 DOI: 10.1007/s12032-024-02429-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
This comprehensive review delves into the multifaceted aspects of genipin, a bioactive compound derived from medicinal plants, focusing on its anti-cancer potential. The review begins by detailing the sources and phytochemical properties of genipin, underscoring its significance in traditional medicine and its transition into contemporary cancer research. It then explores the intricate relationship between genipin's chemical structure and its observed anti-cancer activity, highlighting the molecular underpinnings contributing to its therapeutic potential. This is complemented by a thorough analysis of preclinical studies, which investigates genipin's efficacy against various cancer cell lines and its mechanisms of action at the cellular level. A crucial component of the review is the examination of genipin's bioavailability and pharmacokinetics, providing insights into how the compound is absorbed, distributed, metabolized, and excreted in the body. Then, this review offers a general and updated overview of the anti-cancer studies of genipin and its derivatives based on its basic molecular mechanisms, induction of apoptosis, inhibition of cell proliferation, and disruption of cancer cell signaling pathways. We include information that complements the genipin study, such as toxicity data, and we differentiate this review by including commercial status, disposition, and regulation. Also, this review of genipin stands out for incorporating information on proposals for a technological approach through its load in nanotechnology to improve its bioavailability. The culmination of this information positions genipin as a promising candidate for developing novel anti-cancer drugs capable of supplementing or enhancing current cancer therapies.
Collapse
Affiliation(s)
- Lapava Natallia
- Medicine Standardization Department of Vitebsk State Medical University, Vitebsk, Republic of Belarus.
| | - Aida Dama
- Department of Pharmacy, Faculty of Medical Sciences, Albanian University, Zogu I Blvd., 1001, Tirana, Albania
| | - Era Gorica
- Department of Pharmacy, Faculty of Medical Sciences, Albanian University, Zogu I Blvd., 1001, Tirana, Albania
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, 8952, Schlieren, Zürich, Switzerland
| | - Karaliova Darya
- Medicine Standardization Department of Vitebsk State Medical University, Vitebsk, Republic of Belarus
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de Mexico, Mexico
| | - Antonello Santini
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131, Napoli, Italy.
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico.
| | - Javad Sharifi-Rad
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
69
|
Andoh V, Ocansey DKW, Naveed H, Wang N, Chen L, Chen K, Mao F. The Advancing Role of Nanocomposites in Cancer Diagnosis and Treatment. Int J Nanomedicine 2024; 19:6099-6126. [PMID: 38911500 PMCID: PMC11194004 DOI: 10.2147/ijn.s471360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024] Open
Abstract
The relentless pursuit of effective cancer diagnosis and treatment strategies has led to the rapidly expanding field of nanotechnology, with a specific focus on nanocomposites. Nanocomposites, a combination of nanomaterials with diverse properties, have emerged as versatile tools in oncology, offering multifunctional platforms for targeted delivery, imaging, and therapeutic interventions. Nanocomposites exhibit great potential for early detection and accurate imaging in cancer diagnosis. Integrating various imaging modalities, such as magnetic resonance imaging (MRI), computed tomography (CT), and fluorescence imaging, into nanocomposites enables the development of contrast agents with enhanced sensitivity and specificity. Moreover, functionalizing nanocomposites with targeting ligands ensures selective accumulation in tumor tissues, facilitating precise imaging and diagnostic accuracy. On the therapeutic front, nanocomposites have revolutionized cancer treatment by overcoming traditional challenges associated with drug delivery. The controlled release of therapeutic agents from nanocomposite carriers enhances drug bioavailability, reduces systemic toxicity, and improves overall treatment efficacy. Additionally, the integration of stimuli-responsive components within nanocomposites enables site-specific drug release triggered by the unique microenvironment of the tumor. Despite the remarkable progress in the field, challenges such as biocompatibility, scalability, and long-term safety profiles remain. This article provides a comprehensive overview of recent developments, challenges, and prospects, emphasizing the transformative potential of nanocomposites in revolutionizing the landscape of cancer diagnostics and therapeutics. In Conclusion, integrating nanocomposites in cancer diagnosis and treatment heralds a new era for precision medicine.
Collapse
Affiliation(s)
- Vivian Andoh
- School of Life Sciences, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Dickson Kofi Wiredu Ocansey
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, People’s Republic of China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Central Region, CC0959347, Ghana
| | - Hassan Naveed
- School of Life Sciences, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Naijian Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Liang Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Keping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Fei Mao
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, People’s Republic of China
| |
Collapse
|
70
|
Garg P, Pareek S, Kulkarni P, Salgia R, Singhal SS. Nanoengineering Solutions for Cancer Therapy: Bridging the Gap between Clinical Practice and Translational Research. J Clin Med 2024; 13:3466. [PMID: 38929995 PMCID: PMC11204592 DOI: 10.3390/jcm13123466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Nanoengineering has emerged as a progressive method in cancer treatment, offering precise and targeted delivery of therapeutic agents while concurrently reducing overall toxicity. This scholarly article delves into the innovative strategies and advancements in nanoengineering that bridge the gap between clinical practice and research in the field of cancer treatment. Various nanoengineered platforms such as nanoparticles, liposomes, and dendrimers are scrutinized for their capacity to encapsulate drugs, augment drug efficacy, and enhance pharmacokinetics. Moreover, the article investigates research breakthroughs that drive the progression and enhancement of nanoengineered remedies, encompassing the identification of biomarkers, establishment of preclinical models, and advancement of biomaterials, all of which are imperative for translating laboratory findings into practical medical interventions. Furthermore, the integration of nanotechnology with imaging modalities, which amplify cancer detection, treatment monitoring, and response assessment, is thoroughly examined. Finally, the obstacles and prospective directions in nanoengineering, including regulatory challenges and issues related to scalability, are examined. This underscores the significance of fostering collaboration among various entities in order to efficiently translate nanoengineered interventions into enhanced cancer therapies and patient management.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Siddhika Pareek
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
71
|
Liu Y, Li H, Dai D, He J, Liang Z. Gene Regulatory Mechanism of Mycobacterium Tuberculosis during Dormancy. Curr Issues Mol Biol 2024; 46:5825-5844. [PMID: 38921019 PMCID: PMC11203133 DOI: 10.3390/cimb46060348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) complex, is a zoonotic disease that remains one of the leading causes of death worldwide. Latent tuberculosis infection reactivation is a challenging obstacle to eradicating TB globally. Understanding the gene regulatory network of Mtb during dormancy is important. This review discusses up-to-date information about TB gene regulatory networks during dormancy, focusing on the regulation of lipid and energy metabolism, dormancy survival regulator (DosR), White B-like (Wbl) family, Toxin-Antitoxin (TA) systems, sigma factors, and MprAB. We outline the progress in vaccine and drug development associated with Mtb dormancy.
Collapse
Affiliation(s)
- Yiduo Liu
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Han Li
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Dejia Dai
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| | - Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, No. 100 University West Road, Nanning 530004, China (D.D.)
| |
Collapse
|
72
|
Wang H, Bo W, Feng X, Zhang J, Li G, Chen Y. Strategies and Recent Advances on Improving Efficient Antitumor of Lenvatinib Based on Nanoparticle Delivery System. Int J Nanomedicine 2024; 19:5581-5603. [PMID: 38882543 PMCID: PMC11177867 DOI: 10.2147/ijn.s460844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Lenvatinib (LVN) is a potentially effective multiple-targeted receptor tyrosine kinase inhibitor approved for treating hepatocellular carcinoma, metastatic renal cell carcinoma and thyroid cancer. Nonetheless, poor pharmacokinetic properties including poor water solubility and rapid metabolic, complex tumor microenvironment, and drug resistance have impeded its satisfactory therapeutic efficacy. This article comprehensively reviews the uses of nanotechnology in LVN to improve antitumor effects. With the characteristic of high modifiability and loading capacity of the nano-drug delivery system, an active targeting approach, controllable drug release, and biomimetic strategies have been devised to deliver LVN to target tumors in sequence, compensating for the lack of passive targeting. The existing applications and advances of LVN in improving therapeutic efficacy include improving longer-term efficiency, achieving higher efficiency, combination therapy, tracking and diagnosing application and reducing toxicity. Therefore, using multiple strategies combined with photothermal, photodynamic, and immunoregulatory therapies potentially overcomes multi-drug resistance, regulates unfavorable tumor microenvironment, and yields higher synergistic antitumor effects. In brief, the nano-LVN delivery system has brought light to the war against cancer while at the same time improving the antitumor effect. More intelligent and multifunctional nanoparticles should be investigated and further converted into clinical applications in the future.
Collapse
Affiliation(s)
- Haiqing Wang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Wentao Bo
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Xielin Feng
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Jinliang Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Ge Li
- Department of Emergency, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Yan Chen
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| |
Collapse
|
73
|
Pei J, Natarajan PM, Umapathy VR, Swamikannu B, Sivaraman NM, Krishnasamy L, Palanisamy CP. Advancements in the Synthesis and Functionalization of Zinc Oxide-Based Nanomaterials for Enhanced Oral Cancer Therapy. Molecules 2024; 29:2706. [PMID: 38893579 PMCID: PMC11173400 DOI: 10.3390/molecules29112706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The fabrication of zinc oxide-based nanomaterials (including natural and synthetic polymers like sulfated polysaccharide, chitosan, and polymethyl methacrylate) has potential to improve oral cancer treatment strategies. This comprehensive review explores the diverse synthesis methods employed to fabricate zinc oxide nanomaterials tailored for oral cancer applications. Several synthesis processes, particularly sol-gel, hydrothermal, and chemical vapor deposition approaches, are thoroughly studied, highlighting their advantages and limitations. The review also examines how synthesis parameters, such as precursor selection, the reaction temperature, and growth conditions, influence both the physicochemical attributes and biological efficacy of the resulting nanomaterials. Furthermore, recent advancements in surface functionalization and modification strategies targeted at improving the targeting specificity and pharmaceutical effectiveness of zinc oxide-based nanomaterials in oral cancer therapy are elucidated. Additionally, the review provides insights into the existing issues and prospective views in the field, emphasizing the need for further research to optimize synthesis methodologies and elucidate the mechanisms underlying the efficacy of zinc oxide-based nanoparticles in oral cancer therapy.
Collapse
Affiliation(s)
- Jinjin Pei
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, 2011 QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C., Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Shaanxi University of Technology, Hanzhong 723001, China;
| | - Prabhu Manickam Natarajan
- Department of Clinical Sciences, d Centre of Medical and Bio-Allied Health Sciences and Research, College of Dentistry, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Thai Moogambigai Dental College and Hospital, Chennai 600 107, Tamil Nadu, India;
| | - Bhuminathan Swamikannu
- Department of Prosthodontics, Sree Balaji Dental College and Hospital, Pallikaranai, Chennai 600 100, Tamil Nadu, India;
| | - Nandini Manickam Sivaraman
- Department of Microbiology, Sree Balaji Medical College and Hospital, Bharath University, Chennai 600 100, Tamil Nadu, India; (N.M.S.); (L.K.)
| | - Lakshmi Krishnasamy
- Department of Microbiology, Sree Balaji Medical College and Hospital, Bharath University, Chennai 600 100, Tamil Nadu, India; (N.M.S.); (L.K.)
| | - Chella Perumal Palanisamy
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
74
|
Daneshmehr M, Pazhang M, Mollaei S, Ebadi M, Pazhang Y. Targeted delivery of 5-fluorouracil and shikonin by blended and coated chitosan/pectin nanoparticles for treatment of colon cancer. Int J Biol Macromol 2024; 270:132413. [PMID: 38761911 DOI: 10.1016/j.ijbiomac.2024.132413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
Herein, 5-fluorouracil and shikonin (extracted from Fusarium tricinctum) were loaded in chitosan/pectin nanoparticle (CS/PEC-NPs), prepared by blending (B-CS/PEC-NPs) and coating (C-CS/PEC-NPs) methods. The nanoparticles characterized by Fourier Transform Infrared (FTIR), X-ray diffraction (XRD), Energy-dispersive X-ray (EDX), Scanning Electron Microscope (SEM) and Differential Light Scattering (DLS). Then, some properties of the nanoparticles such as drug release rate and the nanoparticles cytotoxicity were studied. The FTIR, XRD, EDX, SEM and DLS results showed that the nanoparticles synthesized properly with an almost spherical morphology, an average size of 82-93 nm for B-CS/PEC-NPs, an average diameter of below 100 nm (mostly 66-89 nm) for C-CS/PEC-NPs, and hydrodynamic diameter of 310-817 nm. The drug release results indicated the lower release rate of drugs for B-CS/PEC-NPs relative to C-CS/PEC-NPs at different pHs, high release rate of drugs for the nanoparticles in the simulated large intestinal fluids containing pectinase, and Korsmeyer-Peppas model for release of the drugs. The results showed more cytotoxicity of B-CS/PEC-NPs containing drugs, especially B-CS/PEC-NPs containing both drugs (B-CS/PEC/5-FU/SHK-NPs) after treating with pectinase (IC50 of 18.6 μg/mL). In conclusion, despite the limitation of C-CS/PEC-NPs for simultaneous loading of hydrophilic and hydrophobic drugs, B-CS/PEC-NPs showed suitable potency for loading and targeted delivery of the drugs.
Collapse
Affiliation(s)
- Maryam Daneshmehr
- Department of Biology, Faculty of Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Mohammad Pazhang
- Department of Biology, Faculty of Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran.
| | - Saeed Mollaei
- Department of Chemistry, Faculty of Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Mostafa Ebadi
- Department of Biology, Faculty of Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Yaghub Pazhang
- Department of Biology, Faculty of Sciences, Urmia University, Urmia, Iran
| |
Collapse
|
75
|
Amin H, Ibrahim IM, Hassanein EHM. Weaponizing chitosan and its derivatives in the battle against lung cancer. Int J Biol Macromol 2024; 272:132888. [PMID: 38844273 DOI: 10.1016/j.ijbiomac.2024.132888] [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: 12/27/2023] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
Lung cancer (LC) is a crisis of catastrophic proportions. It is a global problem and urgently requires a solution. The classic chemo drugs are lagging behind as they lack selectivity, where their side effects are spilled all over the body, and these adverse effects would be terribly tragic for LC patients. Therefore, they could make a bad situation worse, inflict damage on normal cells, and inflict pain on patients. Since our confidence in classic drugs is eroding, chitosan can offer a major leap forward in LC therapy. It can provide the backbone and the vehicle that enable chemo drugs to penetrate the hard shell of LC. It could be functionalized in a variety of ways to deliver a deadly payload of toxins to kill the bad guys. It is implemented in formulation of polymeric NPs, lipidic NPs, nanocomposites, multiwalled carbon nanotubes, and phototherapeutic agents. This review is a pretty clear proof of chitosan's utility as a weapon in battling LC. Chitosan-based formulations could work effectively to kill LC cells. If a researcher is looking for a vehicle for medication for LC therapy, chitosan can be an appropriate choice.
Collapse
Affiliation(s)
- Haitham Amin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| |
Collapse
|
76
|
Mendes BB, Zhang Z, Conniot J, Sousa DP, Ravasco JMJM, Onweller LA, Lorenc A, Rodrigues T, Reker D, Conde J. A large-scale machine learning analysis of inorganic nanoparticles in preclinical cancer research. NATURE NANOTECHNOLOGY 2024; 19:867-878. [PMID: 38750164 DOI: 10.1038/s41565-024-01673-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 04/10/2024] [Indexed: 06/21/2024]
Abstract
Owing to their distinct physical and chemical properties, inorganic nanoparticles (NPs) have shown promising results in preclinical cancer therapy, but designing and engineering them for effective therapeutic purposes remains a challenge. Although a comprehensive database of inorganic NP research is not currently available, it is crucial for developing effective cancer therapies. In this context, machine learning (ML) has emerged as a transformative tool, but its adaptation to nanomedicine is hindered by inexistent or small datasets. Here we assembled a large database of inorganic NPs, comprising experimental datasets from 745 preclinical studies in cancer nanomedicine. Using descriptive statistics and explainable ML models we mined this database to gain knowledge of inorganic NP design patterns and inform future NP research for cancer treatment. Our analyses suggest that NP shape and therapy type are prominent features in determining in vivo efficacy, measured as a percentage of tumour reduction. Moreover, our database provides a large-scale open-access resource for discriminative ML that the broader nanotechnology community can utilize. Our work blueprints data mining for translational cancer research and offers evidence for standardizing NP reporting to accelerate and de-risk inorganic NP-based drug delivery, which may help to improve patient outcomes in clinical settings.
Collapse
Affiliation(s)
- Bárbara B Mendes
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Zilu Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - João Conniot
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Diana P Sousa
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - João M J M Ravasco
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Lauren A Onweller
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Andżelika Lorenc
- Instituto de Investigação do Medicamento (iMed), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Biopharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Tiago Rodrigues
- Instituto de Investigação do Medicamento (iMed), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal.
| | - Daniel Reker
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA.
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal.
| |
Collapse
|
77
|
Hui San S, Ching Ngai S. E-cadherin re-expression: Its potential in combating TRAIL resistance and reversing epithelial-to-mesenchymal transition. Gene 2024; 909:148293. [PMID: 38373660 DOI: 10.1016/j.gene.2024.148293] [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: 10/13/2023] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 02/21/2024]
Abstract
The major limitation of conventional chemotherapy drugs is their lack of specificity for cancer cells. As a selective apoptosis-inducing agent, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has emerged as an attractive alternative. However, most of the cancer cells are found to be either intrinsically resistant to the TRAIL protein or may develop resistance after multiple treatments, and TRAIL resistance can induce epithelial-to-mesenchymal transition (EMT) at a later stage, promoting cancer invasion and migration. Interestingly, E-cadherin loss has been linked to TRAIL resistance and initiation of EMT, making E-cadherin re-expression a potential target to overcome these obstacles. Recent research suggests that re-expressing E-cadherin may reduce TRAIL resistance by enhancing TRAIL-induced apoptosis and preventing EMT by modulating EMT signalling factors. This reversal of EMT, can also aid in improving TRAIL-induced apoptosis. Therefore, this review provides remarkable insights into the mechanisms underlying E-cadherin re-expression, clinical implications, and potentiation, as well as the research gaps of E-cadherin re-expression in the current cancer treatment.
Collapse
Affiliation(s)
- Ser Hui San
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
78
|
Hristova-Panusheva K, Xenodochidis C, Georgieva M, Krasteva N. Nanoparticle-Mediated Drug Delivery Systems for Precision Targeting in Oncology. Pharmaceuticals (Basel) 2024; 17:677. [PMID: 38931344 PMCID: PMC11206252 DOI: 10.3390/ph17060677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Nanotechnology has emerged as a transformative force in oncology, facilitating advancements in site-specific cancer therapy and personalized oncomedicine. The development of nanomedicines explicitly targeted to cancer cells represents a pivotal breakthrough, allowing the development of precise interventions. These cancer-cell-targeted nanomedicines operate within the intricate milieu of the tumour microenvironment, further enhancing their therapeutic efficacy. This comprehensive review provides a contemporary perspective on precision cancer medicine and underscores the critical role of nanotechnology in advancing site-specific cancer therapy and personalized oncomedicine. It explores the categorization of nanoparticle types, distinguishing between organic and inorganic variants, and examines their significance in the targeted delivery of anticancer drugs. Current insights into the strategies for developing actively targeted nanomedicines across various cancer types are also provided, thus addressing relevant challenges associated with drug delivery barriers. Promising future directions in personalized cancer nanomedicine approaches are delivered, emphasising the imperative for continued optimization of nanocarriers in precision cancer medicine. The discussion underscores translational research's need to enhance cancer patients' outcomes by refining nanocarrier technologies in nanotechnology-driven, site-specific cancer therapy.
Collapse
Affiliation(s)
- Kamelia Hristova-Panusheva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (K.H.-P.); (C.X.)
| | - Charilaos Xenodochidis
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (K.H.-P.); (C.X.)
| | - Milena Georgieva
- Institute of Molecular Biology “Acad. R. Tsanev”, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria;
| | - Natalia Krasteva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, “Acad. Georgi Bonchev” Str., Bl. 21, 1113 Sofia, Bulgaria; (K.H.-P.); (C.X.)
| |
Collapse
|
79
|
Gatto MS, Johnson MP, Najahi-Missaoui W. Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges. Life (Basel) 2024; 14:672. [PMID: 38929656 PMCID: PMC11204409 DOI: 10.3390/life14060672] [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: 03/30/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
In drug development, it is not uncommon that an active substance exhibits efficacy in vitro but lacks the ability to specifically reach its target in vivo. As a result, targeted drug delivery has become a primary focus in the pharmaceutical sciences. Since the approval of Doxil® in 1995, liposomes have emerged as a leading nanoparticle in targeted drug delivery. Their low immunogenicity, high versatility, and well-documented efficacy have led to their clinical use against a wide variety of diseases. That being said, every disease is accompanied by a unique set of physiological conditions, and each liposomal product must be formulated with this consideration. There are a multitude of different targeting techniques for liposomes that can be employed depending on the application. Passive techniques such as PEGylation or the enhanced permeation and retention effect can improve general pharmacokinetics, while active techniques such as conjugating targeting molecules to the liposome surface may bring even further specificity. This review aims to summarize the current strategies for targeted liposomes in the treatment of diseases.
Collapse
Affiliation(s)
| | | | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA; (M.S.G.); (M.P.J.)
| |
Collapse
|
80
|
Alomar TS, AlMasoud N, Awad MA, AlOmar RS, Merghani NM, El-Zaidy M, Bhattarai A. Designing Green Synthesis-Based Silver Nanoparticles for Antimicrobial Theranostics and Cancer Invasion Prevention. Int J Nanomedicine 2024; 19:4451-4464. [PMID: 38799694 PMCID: PMC11127651 DOI: 10.2147/ijn.s440847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/09/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Researchers are increasingly favouring the use of biological resources in the synthesis of metallic nanoparticles. This synthesis process is quick and affordable. The current study examined the antibacterial and anticancer effects of silver nanoparticles (AgNPs) derived from the Neurada procumbens plant. Biomolecules derived from natural sources can be used to coat AgNPs to make them biocompatible. Methods UV-Vis spectroscopy was used to verify the synthesis of AgNPs from Neurada procumbens plant extract, while transmission electron microscopy (TEM), photoluminescence (PL) spectroscopy, dynamic light scattering (DLS), and Fourier transform infrared spectroscopy (FTIR) were used to characterize their morphology, crystalline structure, stability, and coating. Results UV-visible spectrum of AgNPs shows an absorption peak at 422 nm, indicating the isotropic nature of these nanoparticles. As a result of the emergence of a transmission peak at 804.53 and 615.95 cm-1 in the spectrum of the infrared light emitted by atoms in a sample, FTIR spectroscopy demonstrated that the Ag stretching vibration mode is metal-oxygen (M-O). Electron dispersive X-ray (EDX) spectral analysis shows that elementary silver has a peak at 3 keV. Irradiating the silver surface with electrons, photons, or laser beams triggers the illumination. The emission peak locations have been found between 300 and 550 nm. As a result of DLS analysis, suspended particles showed a bimodal size distribution, with their Z-average particle size being 93.38 nm. Conclusion The findings showed that the antibacterial action of AgNPs was substantially (p≤0.05) more evident against Gramme-positive strains (S. aureus and B. cereus) than E. coli. The biosynthesis of AgNPs is an environmentally friendly method for making nanostructures that have antimicrobial and anticancer properties.
Collapse
Affiliation(s)
- Taghrid S Alomar
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Najla AlMasoud
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Manal A Awad
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Reem S AlOmar
- Department of Family and Community Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, 32210, Saudi Arabia
| | - Nada M Merghani
- Central Research Laboratory, Vice Rectorate for Studies and Scientific Research, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohamed El-Zaidy
- Department of Botany and Microbiology, Faculty of Science, King Saud University, Riyadh, 11459, Saudi Arabia
| | - Ajaya Bhattarai
- Department of Chemistry, Mahendra Morang Adarsh Multiple Campus, Tribhuvan University, Biratnagar, 56613, Nepal
| |
Collapse
|
81
|
Lang X, Wang X, Han M, Guo Y. Nanoparticle-Mediated Synergistic Chemoimmunotherapy for Cancer Treatment. Int J Nanomedicine 2024; 19:4533-4568. [PMID: 38799699 PMCID: PMC11127654 DOI: 10.2147/ijn.s455213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Until now, there has been a lack of effective strategies for cancer treatment. Immunotherapy has high potential in treating several cancers but its efficacy is limited as a monotherapy. Chemoimmunotherapy (CIT) holds promise to be widely used in cancer treatment. Therefore, identifying their involvement and potential synergy in CIT approaches is decisive. Nano-based drug delivery systems (NDDSs) are ideal delivery systems because they can simultaneously target immune cells and cancer cells, promoting drug accumulation, and reducing the toxicity of the drug. In this review, we first introduce five current immunotherapies, including immune checkpoint blocking (ICB), adoptive cell transfer therapy (ACT), cancer vaccines, oncolytic virus therapy (OVT) and cytokine therapy. Subsequently, the immunomodulatory effects of chemotherapy by inducing immunogenic cell death (ICD), promoting tumor killer cell infiltration, down-regulating immunosuppressive cells, and inhibiting immune checkpoints have been described. Finally, the NDDSs-mediated collaborative drug delivery systems have been introduced in detail, and the development of NDDSs-mediated CIT nanoparticles has been prospected.
Collapse
Affiliation(s)
- Xiaoxue Lang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, People’s Republic of China
| |
Collapse
|
82
|
Bhuia MS, Chowdhury R, Akter MA, Ali MA, Afroz M, Akbor MS, Sonia FA, Mubarak MS, Islam MT. A mechanistic insight into the anticancer potentials of resveratrol: Current perspectives. Phytother Res 2024. [PMID: 38768953 DOI: 10.1002/ptr.8239] [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: 11/11/2023] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
Resveratrol is a widely recognized polyphenolic phytochemical found in various plants and their fruits, such as peanuts, grapes, and berry fruits. It is renowned for its several health advantages. The phytochemical is well known for its anticancer properties, and a substantial amount of clinical evidence has also established its promise as a chemotherapeutic agent. This study focuses on assessing the anticancer properties of resveratrol and gaining insight into the underlying molecular mechanisms. It also evaluates the biopharmaceutical, toxicological characteristics, and clinical utilization of resveratrol to determine its suitability for further development as a reliable anticancer agent. Therefore, the information about preclinical and clinical studies was collected from different electronic databases up-to-date (2018-2023). Findings from this study revealed that resveratrol has potent therapeutic benefits against various cancers involving different molecular mechanisms, such as induction of oxidative stress, cytotoxicity, inhibition of cell migration and invasion, autophagy, arresting of the S phase of the cell cycle, apoptotic, anti-angiogenic, and antiproliferative effects by regulating different molecular pathways including PI3K/AKT, p38/MAPK/ERK, NGFR-AMPK-mTOR, and so on. However, the compound has poor oral bioavailability due to reduced absorption; this limitation is overcome by applying nanotechnology (nanoformulation of resveratrol). Clinical application also showed therapeutic benefits in several types of cancer with no serious adverse effects. We suggest additional extensive studies to further check the efficacy, safety, and long-term hazards. This could involve a larger number of clinical samples to establish the compound as a reliable drug in the treatment of cancer.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Dhaka, Bangladesh
| | - Mst Asma Akter
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md Arman Ali
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Meher Afroz
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md Showkot Akbor
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Fatema Akter Sonia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
83
|
Wu P, Wang X, Yin M, Zhu W, Chen Z, Zhang Y, Jiang Z, Shi L, Zhu Q. ULK1 Mediated Autophagy-Promoting Effects of Rutin-Loaded Chitosan Nanoparticles Contribute to the Activation of NF-κB Signaling Besides Inhibiting EMT in Hep3B Hepatoma Cells. Int J Nanomedicine 2024; 19:4465-4493. [PMID: 38779103 PMCID: PMC11110815 DOI: 10.2147/ijn.s443117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Background Liver cancer remains to be one of the leading causes of cancer worldwide. The treatment options face several challenges and nanomaterials have proven to improve the bioavailability of several drug candidates and their applications in nanomedicine. Specifically, chitosan nanoparticles (CNPs) are extremely biodegradable, pose enhanced biocompatibility and are considered safe for use in medicine. Methods CNPs were synthesized by ionic gelation, loaded with rutin (rCNPs) and characterized by ultraviolet-visible spectroscopy (UV-Vis), Fourier-transform infrared spectroscopy (FTIR), dynamic light scattering (DLS) and transmission electron microscopy (TEM). The rCNPs were tested for their cytotoxic effects on human hepatoma Hep3B cells, and experiments were conducted to determine the mechanism of such effects. Further, the biocompatibility of the rCNPs was tested on L929 fibroblasts, and their hemocompatibility was determined. Results Initially, UV-vis and FTIR analyses indicated the possible loading of rutin on rCNPs. Further, the rutin load was quantitatively measured using Ultra-Performance Liquid Chromatography (UPLC) and the concentration was 88 µg/mL for 0.22 micron filtered rCNPs. The drug loading capacity (LC%) of the rCNPs was observed to be 13.29 ± 0.68%, and encapsulation efficiency (EE%) was 19.55 ± 1.01%. The drug release was pH-responsive as 88.58% of the drug was released after 24 hrs at the lysosomal pH 5.5, whereas 91.44% of the drug was released at physiological pH 7.4 after 102 hrs. The cytotoxic effects were prominent in 0.22 micron filtered samples of 5 mg/mL rutin precursor. The particle size for the rCNPs at this concentration was 144.1 nm and the polydispersity index (PDI) was 0.244, which is deemed to be ideal for tumor targeting. A zeta potential (ζ-potential) value of 16.4 mV indicated rCNPs with good stability. The IC50 value for the cytotoxic effects of rCNPs on human hepatoma Hep3B cells was 9.7 ± 0.19 μg/mL of rutin load. In addition, the increased production of reactive oxygen species (ROS) and changes in mitochondrial membrane potential (MMP) were observed. Gene expression studies indicated that the mechanism for cytotoxic effects of rCNPs on Hep3B cells was due to the activation of Unc-51-like autophagy-activating kinase (ULK1) mediated autophagy and nuclear factor kappa B (NF-κB) signaling besides inhibiting the epithelial-mesenchymal Transition (EMT). In addition, the rCNPs were less toxic on NCTC clone 929 (L929) fibroblasts in comparison to the Hep3B cells and possessed excellent hemocompatibility (less than 2% of hemolysis). Conclusion The synthesized rCNPs were pH-responsive and possessed the physicochemical properties suitable for tumor targeting. The particles were effectively cytotoxic on Hep3B cells in comparison to normal cells and possessed excellent hemocompatibility. The very low hemolytic profile of rCNPs indicates that the drug could be administered intravenously for cancer therapy.
Collapse
Affiliation(s)
- Peng Wu
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xiaoyong Wang
- The People’s Hospital of Rugao, Nantong, People’s Republic of China
| | - Min Yin
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Wenjie Zhu
- Kangda College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zheng Chen
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yang Zhang
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Ziyu Jiang
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People’s Republic of China
| | - Longqing Shi
- Department of Hepatobiliary and Pancreatic Surgery, Third Affiliated Hospital of Soochow University, Jiangsu, People’s Republic of China
| | - Qiang Zhu
- Children’s Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
84
|
Ilieş BD, Yildiz I, Abbas M. Peptide-conjugated Nanoparticle Platforms for Targeted Delivery, Imaging, and Biosensing Applications. Chembiochem 2024; 25:e202300867. [PMID: 38551557 DOI: 10.1002/cbic.202300867] [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: 12/25/2023] [Revised: 03/09/2024] [Indexed: 04/24/2024]
Abstract
Peptides have become an indispensable tool in engineering of multifunctional nanostructure platforms for biomedical applications such as targeted drug and gene delivery, imaging and biosensing. They can be covalently incorporated into a variety of nanoparticles (NPs) including polymers, metallic nanoparticles, and others. Using different bioconjugation techniques, multifunctional peptide-modified NPs can be formulated to produce therapeutical and diagnostic platforms offering high specificity, lower toxicity, biocompatibility, and stimuli responsive behavior. Targeting peptides can direct the nanoparticles into specific tissues for targeted drug and gene delivery and imaging applications due to their specificity towards certain receptors. Furthermore, due to their stimuli-responsive features, they can offer controlled release of therapeutics into desired sites of disease. In addition, peptide-based biosensors and imaging agents can provide non-invasive detection and monitoring of diseases including cancer, infectious diseases, and neurological disorders. In this review, we covered the design and formulation of recent peptide-based NP platforms, as well as their utilization in in vitro and in vivo applications such as targeted drug and gene delivery, targeting, sensing, and imaging applications. In the end, we provided the future outlook to design new peptide conjugated nanomaterials for biomedical applications.
Collapse
Affiliation(s)
- Bogdan Dragoş Ilieş
- Department of Chemistry, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
- Functional Biomaterials Group, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
| | - Ibrahim Yildiz
- Department of Chemistry, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
- Functional Biomaterials Group, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
| | - Manzar Abbas
- Department of Chemistry, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
- Functional Biomaterials Group, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
| |
Collapse
|
85
|
Wang C, Pan J, Chen S, Qiu L, Hu H, Ji L, Wang J, Liu W, Ni X. Polyvinylpyrrolidone Assisted One-Pot Synthesis of Size-Tunable Cocktail Nanodrug for Multifunctional Combat of Cancer. Int J Nanomedicine 2024; 19:4339-4356. [PMID: 38774026 PMCID: PMC11107942 DOI: 10.2147/ijn.s459428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/30/2024] [Indexed: 05/24/2024] Open
Abstract
Background The in vivo barriers and multidrug resistance (MDR) are well recognized as great challenges for the fulfillment of antitumor effects of current drugs, which calls for the development of novel therapeutic agents and innovative drug delivery strategies. Nanodrug (ND) combining multiple drugs with distinct modes of action holes the potential to circumvent these challenges, while the introduction of photothermal therapy (PTT) can give further significantly enhanced efficacy in cancer therapy. However, facile preparation of ND which contains dual drugs and photothermal capability with effective cancer treatment ability has rarely been reported. Methods In this study, we selected curcumin (Cur) and doxorubicin (Dox) as two model drugs for the creation of a cocktail ND (Cur-Dox ND). We utilized polyvinylpyrrolidone (PVP) as a stabilizer and regulator to prepare Cur-Dox ND in a straightforward one-pot method. Results The size of the resulting Cur-Dox ND can be easily adjusted by tuning the charged ratios. It was noted that both loaded drugs in Cur-Dox ND can realize their functions in the same target cell. Especially, the P-glycoprotein inhibition effect of Cur can synergistically cooperate with Dox, leading to enhanced inhibition of 4T1 cancer cells. Furthermore, Cur-Dox ND exhibited pH-responsive dissociation of loaded drugs and a robust photothermal translation capacity to realize multifunctional combat of cancer for photothermal enhanced anticancer performance. We further demonstrated that this effect can also be realized in 3D multicellular model, which possibly attributed to its superior drug penetration as well as photothermal-enhanced cellular uptake and drug release. Conclusion In summary, Cur-Dox ND might be a promising ND for better cancer therapy.
Collapse
Affiliation(s)
- Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Jiaoyang Pan
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Shaoqing Chen
- Department of Radiology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Huaanzi Hu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Li Ji
- Department of Otorhinolaryngology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Wenjia Liu
- Department of Gastroenterology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Xinye Ni
- Department of Radiology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, People’s Republic of China
| |
Collapse
|
86
|
Angelopoulou A. Nanostructured Biomaterials in 3D Tumor Tissue Engineering Scaffolds: Regenerative Medicine and Immunotherapies. Int J Mol Sci 2024; 25:5414. [PMID: 38791452 PMCID: PMC11121067 DOI: 10.3390/ijms25105414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The evaluation of nanostructured biomaterials and medicines is associated with 2D cultures that provide insight into biological mechanisms at the molecular level, while critical aspects of the tumor microenvironment (TME) are provided by the study of animal xenograft models. More realistic models that can histologically reproduce human tumors are provided by tissue engineering methods of co-culturing cells of varied phenotypes to provide 3D tumor spheroids that recapitulate the dynamic TME in 3D matrices. The novel approaches of creating 3D tumor models are combined with tumor tissue engineering (TTE) scaffolds including hydrogels, bioprinted materials, decellularized tissues, fibrous and nanostructured matrices. This review focuses on the use of nanostructured materials in cancer therapy and regeneration, and the development of realistic models for studying TME molecular and immune characteristics. Tissue regeneration is an important aspect of TTE scaffolds used for restoring the normal function of the tissues, while providing cancer treatment. Thus, this article reports recent advancements in the development of 3D TTE models for antitumor drug screening, studying tumor metastasis, and tissue regeneration. Also, this review identifies the significant opportunities of using 3D TTE scaffolds in the evaluation of the immunological mechanisms and processes involved in the application of immunotherapies.
Collapse
Affiliation(s)
- Athina Angelopoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece
| |
Collapse
|
87
|
Gül D, Önal Acet B, Lu Q, Stauber RH, Odabaşı M, Acet Ö. Revolution in Cancer Treatment: How Are Intelligently Designed Nanostructures Changing the Game? Int J Mol Sci 2024; 25:5171. [PMID: 38791209 PMCID: PMC11120744 DOI: 10.3390/ijms25105171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Nanoparticles (NPs) are extremely important tools to overcome the limitations imposed by therapeutic agents and effectively overcome biological barriers. Smart designed/tuned nanostructures can be extremely effective for cancer treatment. The selection and design of nanostructures and the adjustment of size and surface properties are extremely important, especially for some precision treatments and drug delivery (DD). By designing specific methods, an important era can be opened in the biomedical field for personalized and precise treatment. Here, we focus on advances in the selection and design of nanostructures, as well as on how the structure and shape, size, charge, and surface properties of nanostructures in biological fluids (BFs) can be affected. We discussed the applications of specialized nanostructures in the therapy of head and neck cancer (HNC), which is a difficult and aggressive type of cancer to treat, to give an impetus for novel treatment approaches in this field. We also comprehensively touched on the shortcomings, current trends, and future perspectives when using nanostructures in the treatment of cancer.
Collapse
Affiliation(s)
- Désirée Gül
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
| | - Burcu Önal Acet
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
- Chemistry Department, Faculty of Arts and Science, Aksaray University, Aksaray 68100, Turkey;
| | - Qiang Lu
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
| | - Roland H. Stauber
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
| | - Mehmet Odabaşı
- Chemistry Department, Faculty of Arts and Science, Aksaray University, Aksaray 68100, Turkey;
| | - Ömür Acet
- Department of Otorhinolaryngology Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (B.Ö.A.); (Q.L.); (R.H.S.)
- Pharmacy Services Program, Vocational School of Health Science, Tarsus University, Tarsus 33100, Turkey
| |
Collapse
|
88
|
El-Sayed H, Abdelsalam A, Morad MY, Sonbol H, Ibrahim AM, Tawfik E. Phyto-synthesized silver nanoparticles from Sargassum subrepandum: anticancer, antimicrobial, and molluscicidal activities. FRONTIERS IN PLANT SCIENCE 2024; 15:1403753. [PMID: 38779072 PMCID: PMC11110841 DOI: 10.3389/fpls.2024.1403753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
In the realm of nanotechnology, the use of algae to produce nanoparticles is an environmentally friendly, sustainable, and economically viable strategy. In the present study, the brown macroalgae Sargassum subrepandum was utilized to effectively produce silver nanoparticles (AgNPs). Through various characterization techniques, the AgNPs' structural integrity was confirmed. AgNPs exhibited significant antimicrobial activity against Pseudomonas aeruginosa and Fusarium equiseti. AgNPs showed cytotoxic effects on the MCF-7 breast adenocarcinoma cell line with an IC50 of 12.5 µg/ml. Treatment with AgNPs resulted in a marked reduction in cell viability, alongside evident apoptotic and necrotic morphological changes in the cancer cells. Through molecular docking studies, a deeper understanding of the interaction between AgNPs and crucial proteins related to cancer has been achieved, AgNPs showed a promising molluscicidal action on Biomphalaria alexandrina snails, a Schistosoma mansoni intermediate host. The half-lethal dose (LC50) of AgNPs was determined to be 0.84 mg/L. The potential consequences of its administration include potential disruptions to the glycolysis profile, as well as potential impacts on the steroidal hormone's estrogen and testosterone and certain kidney function tests. This study highlights the diverse uses of algae-synthesized AgNPs, ranging from healthcare to environmental management, demonstrating their importance in advancing nano-biotechnological solutions.
Collapse
Affiliation(s)
- Heba El-Sayed
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
| | - Asmaa Abdelsalam
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
| | - Mostafa Y. Morad
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
| | - Hana Sonbol
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amina M. Ibrahim
- Medical Malacology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Eman Tawfik
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
| |
Collapse
|
89
|
Kashyap A, Kumari M, Singh A, Mukherjee K, Maity D. Current development of theragnostic nanoparticles for women's cancer treatment. Biomed Mater 2024; 19:042001. [PMID: 38471150 DOI: 10.1088/1748-605x/ad3311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/12/2024] [Indexed: 03/14/2024]
Abstract
In the biomedical industry, nanoparticles (NPs-exclusively small particles with size ranging from 1-100 nanometres) are recently employed as powerful tools due to their huge potential in sophisticated and enhanced cancer theragnostic (i.e. therapeutics and diagnostics). Cancer is a life-threatening disease caused by carcinogenic agents and mutation in cells, leading to uncontrolled cell growth and harming the body's normal functioning while affecting several factors like low levels of reactive oxygen species, hyperactive antiapoptotic mRNA expression, reduced proapoptotic mRNA expression, damaged DNA repair, and so on. NPs are extensively used in early cancer diagnosis and are functionalized to target receptors overexpressing cancer cells for effective cancer treatment. This review focuses explicitly on how NPs alone and combined with imaging techniques and advanced treatment techniques have been researched against 'women's cancer' such as breast, ovarian, and cervical cancer which are substantially occurring in women. NPs, in combination with numerous imaging techniques (like PET, SPECT, MRI, etc) have been widely explored for cancer imaging and understanding tumor characteristics. Moreover, NPs in combination with various advanced cancer therapeutics (like magnetic hyperthermia, pH responsiveness, photothermal therapy, etc), have been stated to be more targeted and effective therapeutic strategies with negligible side effects. Furthermore, this review will further help to improve treatment outcomes and patient quality of life based on the theragnostic application-based studies of NPs in women's cancer treatment.
Collapse
Affiliation(s)
- Ananya Kashyap
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Madhubala Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Arnika Singh
- Department of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Koel Mukherjee
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India
| | - Dipak Maity
- Integrated Nanosystems Development Institute, Indiana University Indianapolis, IN 46202, United States of America
- Department of Chemistry and Chemical Biology, Indiana University Indianapolis, IN 46202, United States of America
| |
Collapse
|
90
|
Linde C, Chien YT, Chen Z, Mu Q. Nanoparticle-enhanced PD-1/PD-L1 targeted combination therapy for triple negative breast cancer. Front Oncol 2024; 14:1393492. [PMID: 38756653 PMCID: PMC11096478 DOI: 10.3389/fonc.2024.1393492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Breast cancer with triple-negative subtype (TNBC) presents significant challenges with limited treatment options and a poorer prognosis than others. While PD-1/PD-L1 checkpoint inhibitors have shown promise, their efficacy in TNBC remains constrained. In recent years, nanoparticle (NP) technologies offer a novel approach to enhance cancer therapy by optimizing the tumor microenvironment and augmenting chemo- and immunotherapy effects in various preclinical and clinical settings. This review discusses recent investigations in NP strategies for improving PD-1/PD-L1 blockade-based combination therapy for TNBC. Those include single or multi-therapeutic NPs designed to enhance immunogenicity of the tumor, induce immunogenic cell death, and target immunosuppressive elements within the tumor microenvironment. The investigations also include NPs co-loaded with PD-L1 inhibitors and other therapeutic agents, leveraging targeted delivery and synergistic effects to maximize efficacy while minimizing systemic toxicity. Overall, NP approaches represent a promising avenue for enhancing PD-1/PD-L1 checkpoint blockade-based combination therapy in TNBC and encourage further developmental studies.
Collapse
Affiliation(s)
| | | | | | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States
| |
Collapse
|
91
|
Malaiappan S, P T P, Niveditha S. Green Synthesis and Characterization of Zinc Oxide Nanoparticles Using Catharanthus roseus Extract: A Novel Approach. Cureus 2024; 16:e60407. [PMID: 38883108 PMCID: PMC11179740 DOI: 10.7759/cureus.60407] [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: 04/23/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Nanotechnology enables precise manipulation of matter at the molecular level, with nanoparticles offering diverse applications in medicine and beyond. Green synthesis methods, utilizing natural sources like plant extracts, are favored for their eco-friendliness. Zinc oxide (ZnO) nanoparticles are recognized for their ability to combat microbes and reduce inflammation, which holds promise for biomedical applications. Catharanthus roseus, renowned for its medicinal properties, warrants further exploration in oral health management due to its anti-inflammatory and antioxidant attributes. AIM The current study aimed to synthesize Catharanthus roseus-mediated ZnO nanoparticles and to evaluate their anti-inflammatory and antioxidant activity. MATERIALS AND METHODS Catharanthus roseus powder (1 g) was dissolved in distilled water (100 ml), heated at 60°C for 15-20 minutes, and filtered to obtain 20 ml extract. ZnO nanoparticles were synthesized by adding 0.594 g ZnO powder to 50 ml water, mixed with plant extract, and stirred for 72 hours, and the resulting solution was centrifuged. Nanoparticles were collected and analyzed for Fourier-transform infrared spectroscopy (FTIR) using Bruker's Alpha II FTIR spectrometer (Bruker, Billerica, Massachusetts, United States), antioxidant, and anti-inflammatory activities. RESULTS FTIR analysis revealed characteristic peaks indicative of functional groups present in Catharanthus roseus-mediated ZnO nanoparticles, including O-H, N-O, C-O, C=C, and C≡C-H. Anti-inflammatory activity evaluation showed inhibition ranging from 48% to 89%, with the maximum inhibition at 50 μL concentration. Similarly, antioxidant activity ranged from 62% to 88%, with the maximum inhibition also seen at 50 μL concentration. CONCLUSION Both assays effectively showcased the superior anti-inflammatory and antioxidant activity of the Catharanthus roseus-incorporated ZnO nanoparticles extract compared to the control. This suggests their potential as a viable therapeutic agent for further evaluation.
Collapse
Affiliation(s)
- Sankari Malaiappan
- Department of Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Priyangha P T
- Department of Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Sankari Niveditha
- Department of Dentistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
92
|
Tyagi N, Arya RKK, Bisht D, Wadhwa P, Kumar Upadhyay T, Kumar Sethiya N, Jindal DK, Pandey S, Kumar D. Mechanism and potentialities of photothermal and photodynamic therapy of transition metal dichalcogenides (TMDCs) against cancer. LUMINESCENCE 2024; 39:e4770. [PMID: 38751216 DOI: 10.1002/bio.4770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/20/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024]
Abstract
The ultimate goal of nanoparticle-based phototherapy is to suppress tumor growth. Photothermal therapy (PTT) and photothermal photodynamic therapy (PDT) are two types of physicochemical therapy that use light radiation with multiple wavelength ranges in the near-infrared to treat cancer. When a laser is pointed at tissue, photons are taken in the intercellular and intracellular regions, converting photon energy to heat. It has attracted much interest and research in recent years. The advent of transition materials dichalcogenides (TMDCs) is a revolutionary step in PDT/PTT-based cancer therapy. The TMDCs is a multilayer 2D nano-composite. TMDCs contain three atomic layers in which two chalcogens squash in the transition metal. The chalcogen atoms are highly reactive, and the surface characteristics of TMDCs help them to target deep cancer cells. They absorb Near Infrared (NIR), which kills deep cancer cells. In this review, we have discussed the history and mechanism of PDT/PTT and the use of TMDCs and nanoparticle-based systems, which have been practiced for theranostics purposes. We have also discussed PDT/PTT combined with immunotherapy, in which the cancer cell apoptosis is done by activating the immune cells, such as CD8+.
Collapse
Affiliation(s)
- Neha Tyagi
- Department of Pharmaceutical Sciences, Indraprastha Institute of Management & Technology Saharanpur, U.P., India
| | - Rajeshwar Kamal Kant Arya
- Department of Pharmaceutical Sciences, Sir J. C. Bose Technical Campus Bhimtal, Kumaun University, Nainital, Uttarakhand, India
| | - Dheeraj Bisht
- Department of Pharmaceutical Sciences, Sir J. C. Bose Technical Campus Bhimtal, Kumaun University, Nainital, Uttarakhand, India
- Devsthali Vidyapeeth College of Pharmacy (Veer Madho Singh Bhandari Uttarakhand Technical University Dehradun), Rudrapur, Uttarakhand, India
| | - Pankaj Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Tarun Kumar Upadhyay
- Department of Life Sciences, Parul Institute of Applied Sciences & Research and Development Cell, Parul University, Vadodara, Gujarat, India
| | | | - Deepak Kumar Jindal
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology, Hisar, Haryana, India
| | - Sadanand Pandey
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
- Department of Chemistry, College of Natural Science, Yeungnam University, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| |
Collapse
|
93
|
Lulseged BA, Ramaiyer MS, Michel R, Saad EE, Ozpolat B, Borahay MA. The Role of Nanomedicine in Benign Gynecologic Disorders. Molecules 2024; 29:2095. [PMID: 38731586 PMCID: PMC11085148 DOI: 10.3390/molecules29092095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Nanomedicine has revolutionized drug delivery in the last two decades. Nanoparticles appear to be a promising drug delivery platform in the treatment of various gynecological disorders including uterine leiomyoma, endometriosis, polycystic ovarian syndrome (PCOS), and menopause. Nanoparticles are tiny (mean size < 1000 nm), biodegradable, biocompatible, non-toxic, safe, and relatively inexpensive materials commonly used in imaging and the drug delivery of various therapeutics, such as chemotherapeutics, small molecule inhibitors, immune mediators, protein peptides and non-coding RNA. We performed a literature review of published studies to examine the role of nanoparticles in treating uterine leiomyoma, endometriosis, PCOS, and menopause. In uterine leiomyoma, nanoparticles containing 2-methoxyestradiole and simvastatin, promising uterine fibroid treatments, have been effective in significantly inhibiting tumor growth compared to controls in in vivo mouse models with patient-derived leiomyoma xenografts. Nanoparticles have also shown efficacy in delivering magnetic hyperthermia to ablate endometriotic tissue. Moreover, nanoparticles can be used to deliver hormones and have shown efficacy as a mechanism for transdermal hormone replacement therapy in individuals with menopause. In this review, we aim to summarize research findings and report the efficacy of nanoparticles and nanotherapeutics in the treatment of various benign gynecologic conditions.
Collapse
Affiliation(s)
- Bethlehem A. Lulseged
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (B.A.L.); (M.S.R.)
| | - Malini S. Ramaiyer
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (B.A.L.); (M.S.R.)
| | - Rachel Michel
- Department of Population, Family, and Reproductive Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Eslam E. Saad
- Department of Gynecology and Obstetrics, Johns Hopkins University, 720 Rutland Ave, Baltimore, MD 21205, USA;
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Mostafa A. Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University, 720 Rutland Ave, Baltimore, MD 21205, USA;
| |
Collapse
|
94
|
Slama Y, Arcambal A, Septembre-Malaterre A, Morel AL, Pesnel S, Gasque P. Evaluation of core-shell Fe 3O 4@Au nanoparticles as radioenhancer in A549 cell lung cancer model. Heliyon 2024; 10:e29297. [PMID: 38644868 PMCID: PMC11033100 DOI: 10.1016/j.heliyon.2024.e29297] [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: 06/14/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/23/2024] Open
Abstract
In radiotherapy, metallic nanoparticles are of high interest in the fight against cancer for their radiosensitizing effects. This study aimed to evaluate the ability of core-shell Fe3O4@Au nanoparticles to potentiate the irradiation effects on redox-, pro-inflammatory markers, and cell death of A549 human pulmonary cancer cells. The hybrid Fe3O4@Au nanoparticles were synthesized using green chemistry principles by the sonochemistry method. Their characterization by transmission electron microscopy demonstrated an average size of 8 nm and a homogeneous distribution of gold. The decreased hydrodynamic size of these hybrid nanoparticles compared to magnetite (Fe3O4) nanoparticles showed that gold coating significantly reduced the aggregation of Fe3O4 particles. The internalization and accumulation of the Fe3O4@Au nanoparticles within the cells were demonstrated by Prussian Blue staining. The reactive oxygen species (ROS) levels measured by the fluorescent probe DCFH-DA were up-regulated, as well as mRNA expression of SOD, catalase, GPx antioxidant enzymes, redox-dependent transcription factor Nrf2, and ROS-producing enzymes (Nox2 and Nox4), quantified by RT-qPCR. Furthermore, irradiation coupled with Fe3O4@Au nanoparticles increased the expression of canonical pro-inflammatory cytokines and chemokines (TNF-α, IL-1β, IL-6, CXCL8, and CCL5) assessed by RT-qPCR and ELISA. Hybrid nanoparticles did not potentiate the increased DNA damage detected by immunofluorescence following the irradiation. Nevertheless, Fe3O4@Au caused cellular damage, leading to apoptosis through activation of caspase 3/7, secondary necrosis quantified by LDH release, and cell growth arrest evaluated by clonogenic-like assay. This study demonstrated the potential of Fe3O4@Au nanoparticles to potentiate the radiosensitivity of cancerous cells.
Collapse
Affiliation(s)
- Youssef Slama
- Université de La Réunion, Unité de Recherche Etudes Pharmaco-Immunologiques (EPI), CHU de La Réunion, Site Felix Guyon, Allée des Topazes, SC11021, 97400, Saint-Denis, La Réunion, France
- Clinique Sainte-Clotilde, Groupe Clinifutur, 127 Route de Bois de Nèfles, 97400, Saint-Denis, La Réunion, France
| | - Angelique Arcambal
- Université de La Réunion, Unité de Recherche Etudes Pharmaco-Immunologiques (EPI), CHU de La Réunion, Site Felix Guyon, Allée des Topazes, SC11021, 97400, Saint-Denis, La Réunion, France
| | - Axelle Septembre-Malaterre
- Université de La Réunion, Unité de Recherche Etudes Pharmaco-Immunologiques (EPI), CHU de La Réunion, Site Felix Guyon, Allée des Topazes, SC11021, 97400, Saint-Denis, La Réunion, France
| | - Anne-Laure Morel
- Torskal, Nanosciences, 2 Rue Maxime Rivière, 97490 Sainte-Clotilde, La Réunion, France
| | - Sabrina Pesnel
- Torskal, Nanosciences, 2 Rue Maxime Rivière, 97490 Sainte-Clotilde, La Réunion, France
| | - Philippe Gasque
- Université de La Réunion, Unité de Recherche Etudes Pharmaco-Immunologiques (EPI), CHU de La Réunion, Site Felix Guyon, Allée des Topazes, SC11021, 97400, Saint-Denis, La Réunion, France
| |
Collapse
|
95
|
Janani G, Girigoswami A, Deepika B, Udayakumar S, Girigoswami K. Unveiling the Role of Nano-Formulated Red Algae Extract in Cancer Management. Molecules 2024; 29:2077. [PMID: 38731568 PMCID: PMC11085645 DOI: 10.3390/molecules29092077] [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: 03/21/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Cancer is one of the major causes of death, and its negative impact continues to rise globally. Chemotherapy, which is the most common therapy, has several limitations due to its tremendous side effects. Therefore, developing an alternate therapeutic agent with high biocompatibility is indeed needed. The anti-oxidative effects and bioactivities of several different crude extracts of marine algae have been evaluated both in vitro and in vivo. In the present study, we synthesized the aqueous extract (HA) from the marine algae Amphiroa anceps, and then, a liposome was formulated for that extract (NHA). The extracts were characterized using different photophysical tools like dynamic light scattering, UV-visible spectroscopy, FTIR, scanning electron microscopy, and GC-MS analysis. The SEM image revealed a size range of 112-185 nm for NHA and the GC-MS results showed the presence of octadecanoic acid and n-Hexadecanoic acid in the majority. The anticancer activity was studied using A549 cells, and the NHA inhibited the cancer cells dose-dependently, with the highest killing of 92% at 100 μg/mL. The in vivo studies in the zebrafish model showed that neither the HA nor NHA of Amphiroa anceps showed any teratogenic effect. The outcome of our study showed that NHA can be a potential drug candidate for inhibiting cancer with good biocompatibility up to a dose of 100 μg/mL.
Collapse
Affiliation(s)
| | | | | | | | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, Chennai 603103, India; (G.J.); (B.D.); (S.U.)
| |
Collapse
|
96
|
Mathes D, Macedo LB, Pieta TB, Maia BC, Rodrigues OED, Leal JG, Wendt M, Rolim CMB, Mitjans M, Nogueira-Librelotto DR. Co-Delivery of an Innovative Organoselenium Compound and Paclitaxel by pH-Responsive PCL Nanoparticles to Synergistically Overcome Multidrug Resistance in Cancer. Pharmaceutics 2024; 16:590. [PMID: 38794252 PMCID: PMC11124783 DOI: 10.3390/pharmaceutics16050590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
In this study, we designed the association of the organoselenium compound 5'-Seleno-(phenyl)-3'-(ferulic-amido)-thymidine (AFAT-Se), a promising innovative nucleoside analogue, with the antitumor drug paclitaxel, in poly(ε-caprolactone) (PCL)-based nanoparticles (NPs). The nanoprecipitation method was used, adding the lysine-based surfactant, 77KS, as a pH-responsive adjuvant. The physicochemical properties presented by the proposed NPs were consistent with expectations. The co-nanoencapsulation of the bioactive compounds maintained the antioxidant activity of the association and evidenced greater antiproliferative activity in the resistant/MDR tumor cell line NCI/ADR-RES, both in the monolayer/two-dimensional (2D) and in the spheroid/three-dimensional (3D) assays. Hemocompatibility studies indicated the safety of the nanoformulation, corroborating the ability to spare non-tumor 3T3 cells and human mononuclear cells of peripheral blood (PBMCs) from cytotoxic effects, indicating its selectivity for the cancerous cells. Furthermore, the synergistic antiproliferative effect was found for both the association of free compounds and the co-encapsulated formulation. These findings highlight the antitumor potential of combining these bioactives, and the proposed nanoformulation as a potentially safe and effective strategy to overcome multidrug resistance in cancer therapy.
Collapse
Affiliation(s)
- Daniela Mathes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| | - Letícia Bueno Macedo
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Engenharia e Processos Químicos, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil
| | - Taís Baldissera Pieta
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| | - Bianca Costa Maia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| | - Oscar Endrigo Dorneles Rodrigues
- Departamento de Química, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (O.E.D.R.); (J.G.L.); (M.W.)
| | - Julliano Guerin Leal
- Departamento de Química, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (O.E.D.R.); (J.G.L.); (M.W.)
| | - Marcelo Wendt
- Departamento de Química, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (O.E.D.R.); (J.G.L.); (M.W.)
| | - Clarice Madalena Bueno Rolim
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| | - Montserrat Mitjans
- Departament de Bioquimica i Fisiologia, Facultat de Farmacia i Ciències de l’Alimentaciò, Universitat de Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Daniele Rubert Nogueira-Librelotto
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil; (D.M.); (L.B.M.); (B.C.M.); (C.M.B.R.)
- Laboratório de Testes e Ensaios Farmacêuticos In Vitro, Departamento de Farmácia Industrial, Universidade Federal de Santa Maria, Av. Roraima 1000, Santa Maria 97105-900, Brazil;
| |
Collapse
|
97
|
Rodrigues CF, Correia IJ, Moreira AF. Red blood cell membrane-camouflaged gold-core silica shell nanorods for cancer drug delivery and photothermal therapy. Int J Pharm 2024; 655:124007. [PMID: 38493844 DOI: 10.1016/j.ijpharm.2024.124007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/27/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Gold core mesoporous silica shell (AuMSS) nanorods are multifunctional nanomedicines that can act simultaneously as photothermal, drug delivery, and bioimaging agents. Nevertheless, it is reported that once administrated, nanoparticles can be coated with blood proteins, forming a protein corona, that directly impacts on nanomedicines' circulation time, biodistribution, and therapeutic performance. Therefore, it become crucial to develop novel alternatives to improve nanoparticles' half-life in the bloodstream. In this work, Polyethylenimine (PEI) and Red blood cells (RBC)-derived membranes were combined for the first time to functionalize AuMSS nanorods and simultaneously load acridine orange (AO). The obtained results revealed that the RBC-derived membranes promoted the neutralization of the AuMSS' surface charge and consequently improved the colloidal stability and biocompatibility of the nanocarriers. Indeed, the in vitro data revealed that PEI/RBC-derived membranes' functionalization also improved the nanoparticles' cellular internalization and was capable of mitigating the hemolytic effects of AuMSS and AuMSS/PEI nanorods. In turn, the combinatorial chemo-photothermal therapy mediated by AuMSS/PEI/RBC_AO nanorods was able to completely eliminate HeLa cells, contrasting with the less efficient standalone therapies. Such data reinforce the potential of AuMSS nanomaterials to act simultaneously as photothermal and chemotherapeutic agents.
Collapse
Affiliation(s)
- Carolina F Rodrigues
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ilídio J Correia
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; CIEPQPF - Departamento de Engenharia Química, Universidade de Coimbra, Rua Sílvio Lima, 3030-790 Coimbra, Portugal; AEROG-LAETA, Aerospace Sciences Department, Universidade da Beira Interior, Covilhã, Portugal.
| | - André F Moreira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; CPIRN-UDI/IPG - Centro de Potencial e Inovação em Recursos Naturais, Unidade de Investigação para o Desenvolvimento do Interior do Instituto Politécnico da Guarda, Avenida Dr. Francisco de Sá Carneiro, No. 50, 6300-559 Guarda, Portugal.
| |
Collapse
|
98
|
Yin C, Liufu C, Zhu T, Ye S, Jiang J, Wang M, Wang Y, Shi B. Bladder Cancer in Exosomal Perspective: Unraveling New Regulatory Mechanisms. Int J Nanomedicine 2024; 19:3677-3695. [PMID: 38681092 PMCID: PMC11048230 DOI: 10.2147/ijn.s458397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/06/2024] [Indexed: 05/01/2024] Open
Abstract
Bladder cancer, a prevalent malignant neoplasm of the urinary tract, exhibits escalating morbidity and mortality rates. Current diagnosis standards rely on invasive and costly cystoscopy and histopathology, underscoring the urgency for non-invasive, high-throughput, and cost-effective novel diagnostic techniques to ensure timely detection and standardized treatment. Recent years have witnessed the rise of exosome research in bladder cancer studies. Exosomes contain abundant bioactive molecules that can help elucidate the intricate mechanisms underlying bladder cancer pathogenesis and metastasis. Exosomes hold potential as biomarkers for early bladder cancer diagnosis while also serving as targeted drug delivery vehicles to enhance treatment efficacy and mitigate adverse effects. Furthermore, exosome analyses offer insights into the complex molecular signaling networks implicated in bladder cancer progression, revealing novel therapeutic targets. This review provides a comprehensive overview of prevalent exosome isolation techniques and highlights the promising clinical utility of exosomes in both diagnostic and therapeutic applications in bladder cancer management.
Collapse
Affiliation(s)
- Cong Yin
- Department of Urology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
- Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Cen Liufu
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
- Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Tao Zhu
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
- Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Shuai Ye
- Department of Urology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
- Shenzhen University Health Science Center, Shenzhen, People’s Republic of China
| | - Jiahao Jiang
- Department of Urology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
- Clinical College of Anhui Medical University, Shenzhen, People’s Republic of China
| | - Mingxia Wang
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
| | - Yan Wang
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU-HKUST Medical Center, Shenzhen, People’s Republic of China
| | - Bentao Shi
- Department of Urology, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, People’s Republic of China
| |
Collapse
|
99
|
Salve R, Haldar N, Shaikh A, Samanta R, Sengar D, Patra S, Gajbhiye V. MUC1 aptamer-tethered H40-TEPA-PEG nanoconjugates for targeted siRNA-delivery and gene silencing in breast cancer cells. Front Bioeng Biotechnol 2024; 12:1383495. [PMID: 38699430 PMCID: PMC11063312 DOI: 10.3389/fbioe.2024.1383495] [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: 02/07/2024] [Accepted: 03/26/2024] [Indexed: 05/05/2024] Open
Abstract
With a prevalence of 12.5% of all new cancer cases annually, breast cancer stands as the most common form of cancer worldwide. The current therapies utilized for breast cancer are constrained and ineffective in addressing the condition. siRNA-based gene silencing is a promising method for treating breast cancer. We have developed an aptamer-conjugated dendritic multilayered nanoconjugate to treat breast cancer. Initially, we transformed the hydroxyl groups of the hyperbranched bis-MPA polyester dendrimer into carboxylic groups. Subsequently, we linked these carboxylic groups to tetraethylenepentamine to form a positively charged dendrimer. In addition, the mucin-1 (MUC1) aptamer was attached to the dendrimer using a heterobifunctional polyethylene glycol. Characterizing dendrimers involved 1H NMR and dynamic light scattering techniques at every production stage. A gel retardation experiment was conducted to evaluate the successful binding of siRNA with targeted and non-targeted dendrimers. The targeted dendrimers exhibited no harmful effects on the NIH-3T3 fibroblast cells and RBCs, indicating their biocompatible characteristics. Confocal microscopy demonstrated significant higher uptake of targeted dendrimers than non-targeted dendrimers in MCF-7 breast cancer cells. The real-time PCR results demonstrated that the targeted dendrimers exhibited the most pronounced inhibition of the target gene expression compared to the non-targeted dendrimers and lipofectamine-2000. The caspase activation study confirmed the functional effect of survivin silencing by dendrimer, which led to the induction of apoptosis in breast cancer cells. The findings indicated that Mucin-1 targeted hyperbranched bis-MPA polyester dendrimer carrying siRNA could successfully suppress the expression of the target gene in breast cancer cells.
Collapse
Affiliation(s)
- Rajesh Salve
- Nanobioscience Group, Agharkar Research Institute, Pune, India
- Savitribai Phule Pune University, Pune, India
| | - Niladri Haldar
- Nanobioscience Group, Agharkar Research Institute, Pune, India
- Savitribai Phule Pune University, Pune, India
| | - Aazam Shaikh
- Nanobioscience Group, Agharkar Research Institute, Pune, India
- Savitribai Phule Pune University, Pune, India
| | - Rajkumar Samanta
- Nanobioscience Group, Agharkar Research Institute, Pune, India
- Savitribai Phule Pune University, Pune, India
| | - Devyani Sengar
- Nanobioscience Group, Agharkar Research Institute, Pune, India
- Savitribai Phule Pune University, Pune, India
| | - Surajit Patra
- Nanobioscience Group, Agharkar Research Institute, Pune, India
- Savitribai Phule Pune University, Pune, India
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, Pune, India
- Savitribai Phule Pune University, Pune, India
| |
Collapse
|
100
|
Hiba IH, Koh JK, Lai CW, Mousavi SM, Badruddin IA, Hussien M, Wong JP. Polyrhodanine-based nanomaterials for biomedical applications: A review. Heliyon 2024; 10:e28902. [PMID: 38633652 PMCID: PMC11021909 DOI: 10.1016/j.heliyon.2024.e28902] [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: 11/10/2023] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
Rhodanine is a heterocyclic organic compound that has been investigated for its potential biomedical applications, particularly in drug discovery. Rhodanine derivatives have been examined as the medication options for numerous illnesses, including cancer, inflammation, and infectious diseases. Some rhodanine derivatives have also shown promising activity against drug-resistant strains of bacteria and viruses. One of these derivatives is polyrhodanine (PR), a conducting polymer that has gained attention for its biomedical properties. This review article summarises the latest advancements in creating biomaterials based on PR for biosensing, antimicrobial treatments, and anticancer therapies. The distinctive characteristics of PR, such as biocompatibility, biodegradability, and good conductivity, render it an attractive candidate for these applications. The article also explores obstacles and potential future paths for advancing biomaterials made with PR, including synthesis modifications, characterisation techniques, and in vivo evaluation of biocompatibility and efficacy. Overall, as an emerging research topic, this review emphasises the potential of PR as a promising biomaterial for various biomedical applications and provides insights into the contemporary state of research and prospective directions for investigation.
Collapse
Affiliation(s)
- Ibrahim Huzyan Hiba
- Nanotechnology and Catalysis Research Centre (NANOCAT), Institute for Advanced Studies (IAS), University of Malaya (UM), 50603, Kuala Lumpur, Malaysia
| | - Jin Kwei Koh
- Nanotechnology and Catalysis Research Centre (NANOCAT), Institute for Advanced Studies (IAS), University of Malaya (UM), 50603, Kuala Lumpur, Malaysia
| | - Chin Wei Lai
- Nanotechnology and Catalysis Research Centre (NANOCAT), Institute for Advanced Studies (IAS), University of Malaya (UM), 50603, Kuala Lumpur, Malaysia
| | - Seyyed Mojtaba Mousavi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taiwan
| | - Irfan Anjum Badruddin
- Mechanical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Mohamed Hussien
- Department of Chemistry, Faculty of Science, King Khalid University, P.O. Box 9004, Abha, 61413, Saudi Arabia
| | - Jest Phia Wong
- Harper Elite Sdn Bhd, UG-23, PJ Midtown, Jalan Kemajuan, Seksyen 13, 46200, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| |
Collapse
|