1
|
Yadav PK, Chauhan D, Yadav P, Tiwari AK, Sultana N, Gupta D, Mishra K, Gayen JR, Wahajuddin M, Chourasia MK. Nanotechnology Assisted Drug Delivery Strategies for Chemotherapy: Recent Advances and Future Prospects. ACS APPLIED BIO MATERIALS 2025. [PMID: 40318022 DOI: 10.1021/acsabm.5c00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
In pursuit of the treatment of cancer, nanotechnology engineering has emerged as the simplest and most effective means, with the potential to deliver antitumor chemotherapeutics at the targeted site. Employing nanotechnology for drug delivery provides diverse nanosize particles ranging from one to a thousand nanometers. Reduced size improves drug bioavailability by increasing drug diffusion and decreasing the efflux rate. These nanocarriers offer an enormous scope for modification following the chemical and biological properties of both the drug and its disease. Moreover, these nanoformulations assist in targeting pharmaceutically active drug molecules to the desired site and have gained importance in recent years. Their modern use has revolutionized the antitumor action of many therapeutic agents. Higher drug loading efficiency, thermal stability, easy fabrication, low production cost, and large-scale industrial production draw attention to the application of nanotechnology as a better platform for the delivery of drug molecules. Furthermore, the interaction of nanocarrier technology-assisted agents lowers a drug's toxicity and therapeutic dosage, reduces drug tolerance, and enhances active drug concentration in neoplasm tissue, thus decreasing the concentration in healthy tissue. Nanotechnology-based medications are being widely explored and have depicted effective cancer management in vivo and in vitro systems, leading to many clinical trials with promising results. This review summarizes the innovative impact and application of different nanocarriers developed in recent years in cancer therapy. Subsequently, it also describes the essential findings and methodologies and their effects on cancer treatment. Compared with conventional therapy, nanomedicines can significantly improve the therapeutic effectiveness of antitumor drugs. Thus, the adverse effects associated with healthy tissues are decreased, and adverse effects are scaled back through enhanced permeability and retention effects. Lastly, future insights assisting nanotechnology in active therapeutics delivery and their scope in cancer chemotherapeutics have also been discussed.
Collapse
Affiliation(s)
- Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Chauhan
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pooja Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amrendra K Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nazneen Sultana
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Deepak Gupta
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Keerti Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Jiaur R Gayen
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Richmond Road, Bradford BD7 1DP, United Kingdom
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
2
|
Durmaz A, Mizan Kahyaoğlu İ, Can Aytar E, Danışman Kalındemirtaş F, Sert E, Erol-Bozkurt A, Karakuş S. In Vitro, Anti-Colon Cancer Activity of Green Dumbbell-Shaped Rhododendron luteum-Based Carbon Dots. ChemistryOpen 2025; 14:e202400303. [PMID: 39888200 DOI: 10.1002/open.202400303] [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: 08/15/2024] [Revised: 01/08/2025] [Indexed: 02/01/2025] Open
Abstract
Colorectal cancer is the second most common cause of cancer-related deaths worldwide and the third most common cancer overall. In this study, we investigate the anti-colon cancer potential of phytochemically, and thermally synthesised novel green carbon dots based on Rhododendron luteum (RL-CDs). A new synthesis method was used to produce carbon dots obtained from the Rhododendron luteum (RL) plant in an environmentally friendly manner. The green RL were characterized using Fourier-transform infrared spectroscopy (FTIR), UV-Vis spectroscopy, transmission electron microscopy (TEM), and artificial intelligence (AI)-based TEM analysis. The FTIR spectrum showed peaks corresponding to the hydroxyl (-OH) vibration of polyphenols at 3500 cm-1, the C=O vibration of cellulose derivatives at 1728 cm-1, and the C-O stretching of primary alcohol at 1041 cm-1. Two UV absorption peaks at roughly 253 nm (UV-C range), and 320 nm (UV-B range) were observed. The size of the green RL was measured to be less than 50 nm, and its morphology was characterized as dumbbell-shaped through TEM analysis. In-vitro studies were performed with HCT116 colon cancer, MCF-7 breast cancer, and normal HUVEC cells. The results demonstrated that the RL-CDs exhibited selective cytotoxic activity against HCT116 colon cancer cells. The results show that the RL extract stimulates cancer cell death by decreasing the CD44/24 ratio, and increasing apoptotic activity. These observations suggest that green RL-CDs could be an effective anticancer agent in colon cancer therapy, investigating their potential in this direction could be a promising way for future research.
Collapse
Affiliation(s)
- Alper Durmaz
- Ali Nihat Gökyiğit Botanical Garden Application and Research Center, Artvin Çoruh University, 08000, Artvin, Türkiye
| | - İbrahim Mizan Kahyaoğlu
- Department of Chemistry, Faculty of Science, Ondokuz Mayıs University, 55139, Samsun, Türkiye
| | - Erdi Can Aytar
- Department of Horticulture, Faculty of Agriculture, Usak University, 64200, Usak, Türkiye
| | | | - Esra Sert
- Department of Hematology, Istanbul Faculty of Medicine, Istanbul University, 34093, Istanbul, Türkiye
| | - Ayşe Erol-Bozkurt
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, 34093, Istanbul, Türkiye
| | - Selcan Karakuş
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, 34320, Istanbul, Türkiye
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler, 34220, Istanbul, Türkiye
| |
Collapse
|
3
|
Wen Z, Zhang W, Wu W. The latest applications of exosome-mediated drug delivery in anticancer therapies. Colloids Surf B Biointerfaces 2025; 249:114500. [PMID: 39799609 DOI: 10.1016/j.colsurfb.2025.114500] [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: 08/27/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
In recent years, the significant role of anticancer drugs in cancer treatment has garnered considerable attention. However, the application of these drugs is largely limited by their short half-life in blood circulation, low cellular uptake efficiency, and off-target effects. Exosomes, which serve as crucial messengers in intercellular communication, exhibit unique advantages in molecular delivery compared to traditional synthetic carriers, thereby offering new possibilities for modern drug delivery systems. Exosomes possess organotropic functions and are naturally produced by cells, making them promising candidates for natural drug delivery systems with organotropic properties and minimal side effects. These naturally derived carriers can achieve stable, efficient, and selective delivery of anticancer drugs, thereby enhancing the efficacy and potential of anticancer agents in cancer immunotherapy. This review provides a concise overview of the unique characteristics of exosomes related to anticancer drug delivery, strategies for utilizing exosomes as carriers in cancer therapy, and the latest advancements in the field.
Collapse
Affiliation(s)
- Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
4
|
Yadav K, Sahu KK, Sucheta, Minz S, Pradhan M. Unlocking exosome therapeutics: The critical role of pharmacokinetics in clinical applications. Tissue Cell 2025; 93:102749. [PMID: 39904192 DOI: 10.1016/j.tice.2025.102749] [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: 10/17/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Exosomes are microscopic vesicles released by cells that transport various biological materials and play a vital role in intercellular communication. When they are engineered, they serve as efficient delivery systems for therapeutic agents, making it possible to precisely deliver active pharmaceutical ingredients to organs, tissues, and cells. Exosomes' pharmacokinetics, or how they are transported and metabolized inside the body, is affected by several factors, including their source of origination and the proteins in their cell membranes. The pharmacokinetics and mobility of both native and modified exosomes are being observed in living organisms using advanced imaging modalities such as in vitro-in vivo simulation, magnetic resonance imaging, and positron emission tomography. Establishing comprehensive criteria for the investigation of exosomal pharmacokinetic is essential, given its increasing significance in both therapy and diagnostics. To obtain a thorough understanding of exosome intake, distribution, metabolism, and excretion, molecular imaging methods are crucial. The development of industrial processes and therapeutic applications depends on the precise measurement of exosome concentration in biological samples. To ensure a seamless incorporation of exosomes into clinical practice, as their role in therapeutics grows, it is imperative to conduct a complete assessment of their pharmacokinetics. This review provides a brief on how exosome-based research is evolving and the need for pharmacokinetic consideration to realize the full potential of these promising new therapeutic approaches.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai, Chhattisgarh 491024, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 11 122103, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | | |
Collapse
|
5
|
Tiwari P, Shukla RP, Yadav K, Sharma M, Bakshi AK, Panwar D, Singh N, Agarwal N, Mugale MN, Mishra PR. YIGSR Functionalized Hybrid Exosomes Spatially Target Dasatinib to Laminin Receptors for Precision Therapy in Breast Cancer. Adv Healthc Mater 2025; 14:e2402673. [PMID: 39962816 DOI: 10.1002/adhm.202402673] [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: 07/21/2024] [Revised: 12/17/2024] [Indexed: 04/08/2025]
Abstract
In this study, YIGSR-functionalized exosomes (Exo) are engineered and hybridized with lipid polymeric nanoparticles (LPNPs) followed by loading of chemotherapy Dasatinib (DST) to spatially target laminin receptors on tumors. Exo derived from differentiated macrophages are engineered with YIGSR targeting peptides.These YIGSR-Exo are subsequently fused with LPNPs membranes using the freeze-thaw method, resulting in fused hybrid YIGSR-Exo, which are then loaded with DST, creating DST-FuNP@YIGSR-Exo and targeted breast cancer (BC), leading to enhanced mitochondrial membrane potential (54.50 ±5.0%), increased reactive oxygen species (59.50 ± 6.0%), and apoptosis (63 ± 6.5%), ultimately inducing cell death. Further, cellular uptake and receptor blocking studies confirm the binding affinity and interaction of DST-FuNP@YIGSR-Exo with laminin receptors, Intravenous pharmacokinetic analysis of DST-FuNP@YIGSR-Exo reveals a significant improvement in AUC0-∞, with a 20.84-fold increase compared to free DST and a 1.61-fold enhancement over DST-FuNP@Exo. This is further supported by in vivo imaging and demonstrated improved tumor localization. A tumor regression study shows a 6.8-fold reduction in tumors. Tumor tissue-specific IHC for the Ki67 proliferative marker is significantly reduced in the targeted formulation. The potential of DST-FuNP@YIGSR-Exo as an effective carrier for delivering chemotherapeutic drugs, paving the path for the advancement of biologically obtained nanocarriers for targeted breast cancer.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
6
|
Rai N, Marwaha D, Gautam S, Shukla RP, Sharma M, Singh N, Tiwari P, Urandur S, Teja VB, Sanap SN, Yadav K, Bakshi AK, Mishra PR. Intratumoral delivery of Mitomycin C using bio-responsive Gellan Gum Nanogel: In-vitro evaluation and enhanced chemotherapeutic efficacy. Int J Biol Macromol 2025; 302:140306. [PMID: 39864692 DOI: 10.1016/j.ijbiomac.2025.140306] [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: 09/08/2024] [Revised: 01/03/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
Intratumoral drug delivery systems hold immense promise in overcoming the limitations of conventional IV chemotherapy, particularly in enhancing therapeutic efficacy and minimizing systemic side effects. In this study, we introduce a novel redox-responsive intratumoral nanogel system that combines the biocompatibility of natural polysaccharides with the tailored properties of synthetic polymers. The nanogel features a unique cross-linked architecture incorporating redox-sensitive segments, designed to leverage the elevated glutathione levels in the tumor microenvironment for controlled drug release. Synthesis was performed using a microwave-assisted free radical polymerization technique, which facilitated efficient and rapid cross-linking. A Quality by Design strategy was implemented to optimize key parameters, ensuring the nanogel's suitability for intratumoral delivery, including ideal injectability, viscosity, and drug release characteristics. Mitomycin C (MMC), a chemotherapeutic agent effective against hypoxic tumor cells, was efficiently loaded within the cross-linked nanogel. Optimal stability and drug loading were achieved at a 2:1 nanogel/MMC ratio. The nanogel's structure and composition were confirmed using elemental analysis, FTIR, NMR spectroscopy, and XRD. Stability studies demonstrated its robustness in simulated physiological conditions. In vitro evaluations revealed enhanced cellular uptake of the MMC-loaded nanogel, leading to effective cell cycle arrest, mitochondrial membrane potential disruption, and apoptosis, Co-localization studies with Lysotracker Green, a lysosomal marker, revealed that the nanogels were trafficked to lysosomes. Pharmacokinetic analysis showed significantly reduced systemic exposure (lower plasma Cmax) compared to intravenous administration, while biodistribution studies using IVIS imaging demonstrated prolonged retention of the nanogel within tumor tissues. In vivo studies using a 4T1 xenograft mouse model highlighted the superior antitumor efficacy of the intratumoral nanogel system compared to free MMC. The nanogel treatment resulted in significant tumor volume reduction, minimal changes in body weight, and reduced lung metastasis, as confirmed by histological analysis (HE staining). Ki67 and TUNEL assays of tumor tissues further substantiated the nanogel's ability to suppress proliferation and induce apoptosis. These outcomes directly correlate with our goal of using a redox responsive nanogel system to improve localized drug delivery and minimize systemic side effects. This biodegradable, redox-responsive polymer system represents a significant advance in nanomedicine, offering a promising platform for safe and effective localized cancer therapy.
Collapse
Affiliation(s)
- Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sandeep Urandur
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Venkatesh Banala Teja
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sachin Nasik Sanap
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
7
|
Alfawaz Altamimi AS, Arockia Babu M, Afzal M, Bishoyi AK, Roopashree R, Saini S, Sharma RSK, Pathak PK, Chauhan AS, Goyal K, Ali H, Khan NH, Balaraman AK. Exosomes derived from natural killer cells: transforming immunotherapy for aggressive breast cancer. Med Oncol 2025; 42:114. [PMID: 40100465 DOI: 10.1007/s12032-025-02647-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Natural killer cell-derived exosomes (NK-Exos) hold great promise as immune modulators and immunotherapeutics against cancer due to their intrinsically latent anti-tumor effects. They use these nanosized vesicles to deliver cytotoxic molecules, such as perforin, granzymes, and miRNAs, directly to cancer cells to kill them, avoiding immune suppression. NK-Exos has particular efficacy for treating aggressive breast cancer by modulating the TME to activate the immune response and suppress immunosuppressive factors. Bioengineering advances have extended the therapeutic potential of NK-Exos, which permits precise tumor cell targeting and efficient delivery of therapeutic payloads, including small RNAs and chemotherapeutic agents. In engineered NK-Exos, sensitization of cancer cells to apoptosis, reduction of tumor growth, and resistance to drugs have been demonstrated to be highly effective. When combined, NK-Exos synergizes with radiotherapy, chemotherapy, or checkpoint inhibitors, enhancing therapeutic efficacy, and minimizing systemic toxicity. This review emphasizes the critical role of NK-Exos in breast cancer treatment, their integration into combination therapies, and the need for further research to overcome existing limitations and fully realize their clinical potential.
Collapse
Affiliation(s)
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - R S K Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Division of Research and Innovation, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Haider Ali
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, 63000, Cyberjaya, Selangor, Malaysia.
| |
Collapse
|
8
|
Luo X, McAndrews KM, Kalluri R. Natural and Bioengineered Extracellular Vesicles in Diagnosis, Monitoring and Treatment of Cancer. ACS NANO 2025; 19:5871-5896. [PMID: 39869032 PMCID: PMC12002402 DOI: 10.1021/acsnano.4c11630] [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] [Indexed: 01/28/2025]
Abstract
Extracellular vesicles (EVs) are cell derived nanovesicles which are implicated in both physiological and pathological intercellular communication, including the initiation, progression, and metastasis of cancer. The exchange of biomolecules between stromal cells and cancer cells via EVs can provide a window to monitor cancer development in real time for better diagnostic and interventional strategies. In addition, the process of secretion and internalization of EVs by stromal and cancer cells in the tumor microenvironment (TME) can be exploited for delivering therapeutics. EVs have the potential to provide a targeted, biocompatible, and efficient delivery platform for the treatment of cancer and other diseases. Natural as well as engineered EVs as nanomedicine have immense potential for disease intervention. Here, we provide an overview of current knowledge of EVs' function in cancer progression, diagnostic and therapeutic applications for EVs in the cancer setting, as well as current EV engineering strategies.
Collapse
Affiliation(s)
- Xin Luo
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Kathleen M. McAndrews
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
9
|
Zemanek T, Danisovic L, Nicodemou A. Exosomes and solid cancer therapy: where are we now? Med Oncol 2025; 42:77. [PMID: 39961904 PMCID: PMC11832697 DOI: 10.1007/s12032-025-02626-3] [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: 12/21/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Cancer immunotherapy has revolutionized oncology, offering new hope for patients with previously incurable cancers. However, solid tumors remain a significant challenge due to immune evasion, therapeutic resistance, and the immunosuppressive tumor microenvironment. Exosomes, a specialized subset of extracellular vesicles, have emerged as promising tools in cancer therapy owing to their unique role in intercellular communication and immune modulation. These vesicles transport antigens, major histocompatibility complex (MHC) molecules, and immune-modulatory cargo, positioning them as potential platforms for cancer vaccines, drug delivery systems, and combinatorial therapies. Advances in engineered exosomes have improved drug bioavailability, tumor targeting, and immune stimulation, showcasing their potential in personalized medicine. This review highlights their multifaceted role in the tumor microenvironment, and their mechanisms of action in solid cancer therapy. Additionally, we discuss emerging strategies to overcome clinical and technical hurdles, paving the way for novel and effective cancer treatments.
Collapse
Affiliation(s)
- Tomas Zemanek
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- GAMMA - ZA s.r.o., Trencin, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Andreas Nicodemou
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
- GAMMA - ZA s.r.o., Trencin, Slovakia.
| |
Collapse
|
10
|
Sardar A, Rai D, Tripathi AK, Chutani K, Sinha S, Dhaniya G, Trivedi R. FDA-approved polypeptide PTH 1-34 impedes palmitic acid-mediated osteoblasts dysfunction by promoting its differentiation and thereby improving skeletal health. Mol Cell Endocrinol 2025; 597:112445. [PMID: 39719245 DOI: 10.1016/j.mce.2024.112445] [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: 10/03/2024] [Revised: 12/03/2024] [Accepted: 12/20/2024] [Indexed: 12/26/2024]
Abstract
Excessive consumption of saturated fatty acids creates a debilitating cellular environment that hinders the normal function and survival of osteoblasts, contributing to bone metabolic disorders such as osteoporosis. The FDA-approved polypeptide PTH 1-34 is a well-established therapy for post-menopausal osteoporosis, yet its protective effects in a palmitic acid (PA)-rich hyperlipidemic environment are not well understood. This study investigates the impact of PTH 1-34 on PA-induced cellular responses in osteoblasts. Experiments were conducted on mouse and human-derived osteoblasts as well as C57BL/6J male mice. PA was found to suppress osteoblast differentiation, increase apoptosis, and disrupt autophagy, and thereby impair cellular health. Conversely, PTH 1-34 enhanced cellular health by counteracting these effects. At the molecular level, PTH 1-34 exerted its bioactivity by modulating PTH signaling components such as cAMP and CREB. Impaired osteogenic differentiation was restored by modulating bone-anabolic genes. PTH 1-34 also improved mitochondrial health by preserving mitochondrial membrane potential and maintaining the Bax/Bcl2 ratio, thereby improving cellular viability. Additionally, PTH 1-34 regulated autophagic processes, as evidenced by balanced p62 and LC3 levels, further validated using the autophagy inhibitor Bafilomycin A1. In vivo studies in C57BL/6J male mice corroborated these findings. PTH 1-34 reversed the PA action by maintaining osteoblast number and function. This study establishes the protective role of PTH 1-34 in safeguarding osteoblasts from lipotoxicity caused by excessive PA accumulation, highlighting its potential repurposing for patients with lipid-induced skeletal dysfunctions. The new data underscores the therapeutic versatility of the FDA-approved polypeptide PTH 1-34 in managing lipid-related bone health issues.
Collapse
Affiliation(s)
- Anirban Sardar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Divya Rai
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | | | - Kunal Chutani
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shradha Sinha
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Geeta Dhaniya
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
11
|
Li J, Wang J, Chen Z. Emerging role of exosomes in cancer therapy: progress and challenges. Mol Cancer 2025; 24:13. [PMID: 39806451 PMCID: PMC11727182 DOI: 10.1186/s12943-024-02215-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
This review highlights recent progress in exosome-based drug delivery for cancer therapy, covering exosome biogenesis, cargo selection mechanisms, and their application across multiple cancer types. As small extracellular vesicles, exosomes exhibit high biocompatibility and low immunogenicity, making them ideal drug delivery vehicles capable of efficiently targeting cancer cells, minimizing off-target damage and side effects. This review aims to explore the potential of exosomes in cancer therapy, with a focus on applications in chemotherapy, gene therapy, and immunomodulation. Additionally, challenges related to exosome production and standardization are analyzed, highlighting the importance of addressing these issues for their clinical application. In conclusion, exosome-based drug delivery systems offer promising potential for future cancer therapies. Further research should aim to enhance production efficiency and facilitate clinical translation, paving the way for innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Jiale Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China
| | - Jiachong Wang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
| | - Zigui Chen
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
| |
Collapse
|
12
|
Debnath M, Sarkar S, Debnath SK, Dkhar DS, Kumari R, Vaskuri GSSJ, Srivastava A, Chandra P, Prasad R, Srivastava R. Photothermally Active Quantum Dots in Cancer Imaging and Therapeutics: Nanotheranostics Perspective. ACS APPLIED BIO MATERIALS 2024; 7:8126-8148. [PMID: 39526826 DOI: 10.1021/acsabm.4c01190] [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: 11/16/2024]
Abstract
Cancer is becoming a global threat, as the cancerous cells manipulate themselves frequently, resulting in mutants and more abnormalities. Early-stage and real-time detection of cancer biomarkers can provide insight into designing cost-effective diagnostic and therapeutic modalities. Nanoparticle and quantum dot (QD)-based approaches have been recognized as clinically relevant methods to detect disease biomarkers at the molecular level. Over decades, as an emergent noninvasive approach, photothermal therapy has evolved to eradicate cancer. Moreover, various structures, viz., nanoparticles, clusters, quantum dots, etc., have been tested as bioimaging and photothermal agents to identify tumor cells selectively. Among them, QDs have been recognized as versatile probes. They have attracted enormous attention for imaging and therapeutic applications due to their unique colloidal stability, optical and physicochemical properties, biocompatibility, easy surface conjugation, scalable production, etc. However, a few critical concerns of QDs, viz., precise engineering for molecular imaging and sensing, selective interaction with the biological system, and their associated toxicity, restrict their potential intervention in curing cancer and are yet to be explored. According to the U.S. Food and Drug Administration (FDA), there is no specific regulation for the approval of nanomedicines. Therefore, these nanomedicines undergo the traditional drug, biological, and device approval process. However, the market survey of QDs is increasing, and their prospects in translational nanomedicine are very promising. From this perspective, we discuss the importance of QDs for imaging, sensing, and therapeutic usage pertinent to cancer, especially in its early stages. Moreover, we also discuss the rapidly growing translational view of QDs. The long-term safety studies and cellular interaction of these QDs could enhance their visibility and bring photothermally active QDs to the clinical stage and concurrently to FDA approval.
Collapse
Affiliation(s)
- Monalisha Debnath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sayoni Sarkar
- Center for Research in Nanotechnology and Sciences, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sujit Kumar Debnath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Daphika S Dkhar
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rohini Kumari
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | | | - Ananya Srivastava
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rajendra Prasad
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
13
|
Ibrahim M, Fathalla Z, Fatease AA, Alamri AH, Abdelkader H. Breast cancer epidemiology, diagnostic barriers, and contemporary trends in breast nanotheranostics and mechanisms of targeting. Expert Opin Drug Deliv 2024; 21:1735-1754. [PMID: 39361257 DOI: 10.1080/17425247.2024.2412823] [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: 10/23/2023] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
INTRODUCTION Breast cancer is one of the main causes of mortality in women globally. Early and accurate diagnosis represents a milestone in cancer management. Several breast cancer diagnostic agents are available. Many chemotherapeutic agents in conventional dosage forms are approved; nevertheless, they lack cancer cell specificity, resulting in improper treatment and undesirable side effects. Recently, nanotheranostics has emerged as a new paradigm to achieve safe and effective cancer diagnosis and management. AREA COVERED This review provides insight into breast cancer epidemiology, barriers hindering the early diagnosis, and effective delivery of chemotherapeutics. Also, conventional diagnostic agents and recent nanotheranostic platforms have been used in breast cancer. In addition, mechanisms of cancer cell targeting and nano-carrier surface functionalization as an effective approach for chemotherapeutic targeting were reviewed along with future perspectives. EXPERT OPINION We proposed that modified nano-carriers may provide an efficacious approach for breast cancer drug targeting. These nanotheranostics need more clinical evaluations to confirm their efficacy in cancer management. In addition, we recommend the use of artificial intelligence (AI) as a promising approach for early and efficient assessment of breast lesions. AI allows better interpretation and analysis of nanotheranostic data, which minimizes misdiagnosis and avoids the belated intervention of health care providers.
Collapse
Affiliation(s)
- Mohamed Ibrahim
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Zeinab Fathalla
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ali H Alamri
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Hamdy Abdelkader
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
14
|
Saadh MJ, Al-Rihaymee AMA, Kaur M, Kumar A, Mutee AF, Ismaeel GL, Shomurotova S, Alubiady MHS, Hamzah HF, Alhassan ZAA, Alazzawi TS, Muzammil K, Alhadrawi M. Advancements in Exosome Proteins for Breast Cancer Diagnosis and Detection: With a Focus on Nanotechnology. AAPS PharmSciTech 2024; 25:276. [PMID: 39604642 DOI: 10.1208/s12249-024-02983-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024] Open
Abstract
Breast cancer, a leading cause of mortality among women, has been recognized as requiring improved diagnostic methods. Exosome proteins, found in small extracellular vesicles, have emerged as a promising solution, reflecting the state of their cell of origin and playing key roles in cancer progression. This review examines their potential in breast cancer diagnosis, discussing advanced isolation and characterization techniques such as ultracentrifugation and microfluidic-based approaches. Various detection methods-including electrochemical, nano-based, optical, and machine learning platforms-were evaluated for their high sensitivity, specificity, and non-invasive capabilities. Electrochemical methods were used to identify unique protein signatures for rapid, cost-effective diagnosis, while machine learning enhanced the classification of exosome proteins. Nano-based techniques leveraged nanomaterials to detect low-abundance proteins, and optical methods offered real-time, label-free monitoring. Despite their promise, challenges in standardizing protocols and integrating these diagnostics into clinical practice remain. Future directions include technological advancements, personalized medicine, and exploring the therapeutic potential of exosome proteins.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | - Afrah Majeed Ahmed Al-Rihaymee
- Anesthesia Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, Babylon, 51001, Iraq
| | - Mandeep Kaur
- Department of Chemistry, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, 560069, Karnataka, India
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, 247341, Uttar Pradesh, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | | | - Ghufran Lutfi Ismaeel
- Department of Pharmacology, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | - Shirin Shomurotova
- Department of Chemistry Teaching Methods, Tashkent State Pedagogical University named after Nizami, Bunyodkor street 27, Tashkent, Uzbekistan
| | | | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | - Tuqa S Alazzawi
- Collage of Dentist, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Khamis Mushait Campus, Abha, 62561, Saudi Arabia
| | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
15
|
Guan C, Han Y, Ling Z, Meng X, Zhang B, Dong W, Zhang D, Chen K. Nanomaterials: breaking the bottleneck of breast cancer drug resistance. Front Immunol 2024; 15:1492546. [PMID: 39606228 PMCID: PMC11599193 DOI: 10.3389/fimmu.2024.1492546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Drug resistance poses a significant challenge in the treatment of breast cancer. In recent years, a variety of nanomaterials have been discovered and synthesized that can selectively target tumor cells and play a crucial role in the advancement of breast cancer therapies. As our understanding of tumor heterogeneity deepens, the emerging potential of nanomaterials in addressing drug resistance has garnered considerable attention. These materials not only selectively target tumor cells but also possess unique properties that make them promising options for cancer treatment, including low toxicity, excellent biocompatibility, ease of preparation, the ability to carry antitumor drugs, and customizable surface functions. In this review, we will comprehensively summarize two key developments in breast cancer treatment: the application of antitumor drugs and nanomaterials. We will explore the mechanisms by which nanomaterials improve drug resistance in breast cancer, targeted nanotherapy strategies to mitigate this resistance, and recent research advancements in anticancer nanomaterials. This overview aims to highlight the significant role of nanomaterials in breast cancer treatment and provide a theoretical framework for identifying optimal treatment strategies in the future.
Collapse
Affiliation(s)
- Chao Guan
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Yahao Han
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| | - Zhenzheng Ling
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Xiang Meng
- The First Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Baolin Zhang
- The Fourth Clinical College of China Medical University, Shenyang, Liaoning, China
| | - Wanwei Dong
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| | - Di Zhang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Keyan Chen
- Laboratory Animal Science of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
16
|
Yadav K. Nanotechnology in diabetes Management: Revolutionizing treatment and diagnostics. J Mol Liq 2024; 414:126117. [DOI: 10.1016/j.molliq.2024.126117] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Ale Y, Nainwal N. Exosomes as nanocarrier for Neurotherapy: Journey from application to challenges. J Drug Deliv Sci Technol 2024; 101:106312. [DOI: 10.1016/j.jddst.2024.106312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
18
|
Feng M, Zhang L, Zou Z, Xie M, Zhang J, Wang J, Wang K, Zhu J, Xiong L. Engineered Macrophage Exosomes Deliver Drug-Targeted Therapy for Breast Cancer. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2012. [PMID: 39510065 DOI: 10.1002/wnan.2012] [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: 05/14/2024] [Revised: 09/21/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Breast cancer is a highly widespread form of malignant tumor characterized by a high rate of recurrence and mortality; it primarily occurs when tumor cells spread to peripheral regions of the body. Macrophages have a significant impact on the proliferation and metastasis of breast cancer. The exosomes generated by these cells exhibit an extensive spectrum of capabilities in suppressing the spread of cancer cells. These feature very specific targeting properties for breast cancer cells and inhibit the proliferation of cancer cells by altering the immune milieu within the tumor. This study investigates methods for developing macrophage-derived exosomes, such as using protein-coupled exosome membranes to protect delivery contents, creating multifunctional biomimetic particles, and utilizing ultrasonic fusion to protect delivery contents. Furthermore, this paper addresses recent advances in producing macrophage exosomes from organic and inorganic materials. In general, targeted treatment for breast cancer could benefit greatly from creating drug delivery systems mediated by macrophage exosomes.
Collapse
Affiliation(s)
- Mingrui Feng
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Basic Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
- Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Lifang Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Basic Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhuoling Zou
- Queen Mary College, Nanchang University, Nanchang, China
| | - Mengying Xie
- Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Jianbo Zhang
- First Clinical Medical College, Nanchang University, Nanchang, China
| | - Jiayang Wang
- First Clinical Medical College, Nanchang University, Nanchang, China
| | - Keqin Wang
- First Clinical Medical College, Nanchang University, Nanchang, China
| | - Jun Zhu
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Basic Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lixia Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Basic Medical College, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
19
|
Patel D, Solanki J, Kher MM, Azagury A. A Review: Surface Engineering of Lipid-Based Drug Delivery Systems. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401990. [PMID: 39004869 DOI: 10.1002/smll.202401990] [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: 03/13/2024] [Revised: 05/19/2024] [Indexed: 07/16/2024]
Abstract
This review explores the evolution of lipid-based nanoparticles (LBNPs) for drug delivery (DD). Herein, LBNPs are classified into liposomes and cell membrane-based nanoparticles (CMNPs), each with unique advantages and challenges. Conventional LBNPs possess drawbacks such as poor targeting, quick clearance, and limited biocompatibility. One of the possible alternatives to overcome these challenges is surface modification of nanoparticles (NPs) with materials such as polyethylene glycol (PEG), aptamers, antibody fragments, peptides, CD44, hyaluronic acid, folic acid, palmitic acid, and lactoferrin. Thus, the main focus of this review will be on the different surface modifications that enable LBNPs to have beneficial properties for DD, such as enhancing mass transport properties, immune evasion, improved stability, and targeting. Moreover, various CMNPs are explored used for DD derived from cells such as red blood cells (RBCs), platelets, leukocytes, cancer cells, and stem cells, highlighting their unique natural properties (e.g., biocompatibility and ability to evade the immune system). This discussion extends to the biomimicking of hybrid NPs accomplished through the surface coating of synthetic (mainly polymeric) NPs with different cell membranes. This review aims to provide a comprehensive resource for researchers on recent advances in the field of surface modification of LBNPs and CMNPs. Overall, this review provides valuable insights into the dynamic field of lipid-based DD systems.
Collapse
Affiliation(s)
- Dhaval Patel
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel, 4070000, Israel
| | - Jyoti Solanki
- Post Graduate Department of Biosciences, Sardar Patel University, Bakrol, Anand, Gujarat, 388120, India
| | - Mafatlal M Kher
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel, 4070000, Israel
| | - Aharon Azagury
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel, 4070000, Israel
| |
Collapse
|
20
|
Biswas A, Choudhury AD, Mishra A, Verma SK, Bisen AC, Sanap SN, Agrawal S, Kumar M, Kumar S, Bhatta RS. Pharmacokinetic Analysis of Gatifloxacin and Dexamethasone in Rabbit Ocular Biofluid Using a Sensitive and Selective LC-MS/MS Method. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5088. [PMID: 39351617 DOI: 10.1002/jms.5088] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/21/2024] [Indexed: 12/29/2024]
Abstract
Bacterial keratitis (BK) is an infection that causes inflammation of the cornea and, if severe, can result in blindness. Topical fluoroquinolones combined with corticosteroids have been shown to be useful in the treatment of BK. A rapid, selective, and sensitive bioanalytical method for simultaneous quantification of Gatifloxacin (GAT) and Dexamethasone (DEX) has been developed and validated using tandem mass spectrometry (LC-MS/MS). Optimal separation was accomplished in under 5 min using an Agilent Zorbax C18 column (100 mm × 4.6 mm, 3.5 μm). The mobile phase was composed of a blend of 0.2% formic acid in triple distilled water and methanol with a flow rate of 0.65 mL/min in isocratic mode. GAT and DEX were detected in positive electrospray ionization multiple reaction monitoring mode (MRM), and the retention time was found to be at 1.64 and 2.93 min, respectively. The linearity of GAT and DEX was found to be in the range of 1.56-400 ng mL-1 with good precision and accuracy. The method was validated according to USFDA regulatory guidelines. The validated method was effectively utilized for preclinical pharmacokinetic analysis of GAT and DEX in rabbit tear fluid following the topical application of a commercial formulation.
Collapse
Affiliation(s)
- Arpon Biswas
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Pharmaceutics and Pharmacokinetics Division of CSIR-Central Drug Research Institute, Jawaharlal Nehru University, New Delhi, India
| | - Abhijit Deb Choudhury
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Pharmaceutics and Pharmacokinetics Division of CSIR-Central Drug Research Institute, Jawaharlal Nehru University, New Delhi, India
| | - Anjali Mishra
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sarvesh Kumar Verma
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Pharmaceutics and Pharmacokinetics Division of CSIR-Central Drug Research Institute, Jawaharlal Nehru University, New Delhi, India
| | - Amol Chhatrapati Bisen
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sachin Nashik Sanap
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sristi Agrawal
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mukesh Kumar
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Pharmaceutics and Pharmacokinetics Division of CSIR-Central Drug Research Institute, Jawaharlal Nehru University, New Delhi, India
| | - Shivansh Kumar
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rabi Sankar Bhatta
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
21
|
Shukla RP, Tiwari P, Sardar A, Urandur S, Gautam S, Marwaha D, Tripathi AK, Rai N, Trivedi R, Mishra PR. Alendronate-functionalized porous nano-crystalsomes mitigate osteolysis and consequent inhibition of tumor growth in a tibia-induced metastasis model. J Control Release 2024; 372:331-346. [PMID: 38844176 DOI: 10.1016/j.jconrel.2024.06.009] [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: 02/03/2024] [Revised: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
Bone is one of the most prevalent sites of metastases in various epithelial malignancies, including breast cancer and this metastasis to bone often leads to severe skeletal complications in women due to its osteolytic nature. To address this, we devised a novel drug delivery approach using an Alendronate (ALN) functionalized self-assembled porous crystalsomes for concurrent targeting of Oleanolic acid (OA) and ALN (ALN + OA@NCs) to bone metastasis. Initially, the conjugation of both PEG-OA and OA-PEG-ALN with ALN and OA was achieved, and this conjugation was then self-assembled into porous crystalsomes (ALN + OA@NCs) by nanoemulsion crystallization. The reconstruction of a 3D single particle using transmission electron microscopy ensured the crystalline porous structure of ALN + OA@NCs, was well aligned with characteristic nanoparticle attributes including size distribution, polydispersity, and zeta potential. Further, ALN + OA@NCs showed enhanced efficacy in comparison to OA@NCs suggesting the cytotoxic roles of ALN towards cancer cells, followed by augmentation ROS generation (40.81%), mitochondrial membrane depolarization (57.20%), and induction of apoptosis (40.43%). We found that ALN + OA@NCs facilitated inhibiting osteoclastogenesis and bone resorption followed by inhibited osteolysis. In vivo activity of ALN + OA@NCs in the 4 T1 cell-induced tibia model rendered a reduced bone loss in the treated mice followed by restoring bone morphometric markers which were further corroborated bone-targeting effects of ALN + OA@NCs to reduce RANKL-stimulated osteoclastogenesis. Further, In vivo intravenous pharmacokinetics showed the improved therapeutic profile of the ALN + OA@NCs in comparison to the free drug, prolonging the levels of the drug in the systemic compartment by reducing the clearance culminating the higher accumulation at the tumor site. Our finding proposed that ALN + OA@NCs can effectively target and treat breast cancer metastasis to bone and its associated complications.
Collapse
Affiliation(s)
- Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anirban Sardar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sandeep Urandur
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ashish Kumar Tripathi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
22
|
Yadav K, Gnanakani SPE, Sahu KK, Veni Chikkula CK, Vaddi PS, Srilakshmi S, Yadav R, Sucheta, Dubey A, Minz S, Pradhan M. Nano revolution of DNA nanostructures redefining cancer therapeutics-A comprehensive review. Int J Biol Macromol 2024; 274:133244. [PMID: 38901506 DOI: 10.1016/j.ijbiomac.2024.133244] [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: 01/10/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
DNA nanostructures are a promising tool in cancer treatment, offering an innovative way to improve the effectiveness of therapies. These nanostructures can be made solely from DNA or combined with other materials to overcome the limitations of traditional single-drug treatments. There is growing interest in developing nanosystems capable of delivering multiple drugs simultaneously, addressing challenges such as drug resistance. Engineered DNA nanostructures are designed to precisely deliver different drugs to specific locations, enhancing therapeutic effects. By attaching targeting molecules, these nanostructures can recognize and bind to cancer cells, increasing treatment precision. This approach offers tailored solutions for targeted drug delivery, enabling the delivery of multiple drugs in a coordinated manner. This review explores the advancements and applications of DNA nanostructures in cancer treatment, with a focus on targeted drug delivery and multi-drug therapy. It discusses the benefits and current limitations of nanoscale formulations in cancer therapy, categorizing DNA nanostructures into pure forms and hybrid versions optimized for drug delivery. Furthermore, the review examines ongoing research efforts and translational possibilities, along with challenges in clinical integration. By highlighting the advancements in DNA nanostructures, this review aims to underscore their potential in improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai 490024, India
| | - S Princely E Gnanakani
- Department of Pharmaceutical Biotechnology, Parul Institute of Pharmacy, Parul University, Post Limda, Ta.Waghodia - 391760, Dist. Vadodara, Gujarat, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - C Krishna Veni Chikkula
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, USA
| | - Poorna Sai Vaddi
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, USA
| | - S Srilakshmi
- Gitam School of Pharmacy, Department of Pharmaceutical Chemistry, Gitams University, Vishakhapatnam, India
| | - Renu Yadav
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana 122103, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana 122103, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru 575018, Karnataka, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak (M.P.), India
| | | |
Collapse
|
23
|
Kazeminava F, Javanbakht S, Latifi Z, Rasoulzadehzali M, Abbaszadeh M, Alimohammadzadeh B, Mahdipour M, Fattahi A, Hamishehkar H, Adibag Z, Nouri M. Ultrasound-assisted encapsulating folic acid-based carbon quantum dots within breast cancer cell-derived exosomes as a co-receptors-mediated anticancer nanocarrier for enhanced breast cancer therapy. Sci Rep 2024; 14:16941. [PMID: 39043763 PMCID: PMC11266556 DOI: 10.1038/s41598-024-67934-6] [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: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
The nonspecific nature of cancer drug delivery often results in substantial toxic side effects during treatments for breast cancer. To mitigate these negative outcomes, our approach involves loading methotrexate (MTX) within carbon quantum dots (CQDs) synthesized from folic acid, which are then enveloped in exosomal membranes obtained from breast cancer cells (Ex@MTX-CQDs). Analysis utilizing nanoparticle tracking techniques has demonstrated that these Ex@MTX-CQDs maintain the physical and biochemical properties of their exosomal precursors. The release profile of MTX indicated a restricted release percentage (less than 10%) under normal physiological conditions, which is contrasted by a more consistent release rate (approximately 65%) when emulating the conditions found within tumor tissues. The toxicological assessments have confirmed that the presence of exosomes combined with leftover folic acid significantly improves the delivery efficacy of MTX directly to the cancerous cells through the binding to folate and heparan sulfate proteoglycan receptors. This process results in increased disruption of the mitochondrial membrane potential and subsequently triggers apoptosis, ultimately leading to the destruction of cancerous cells. Our research could potentially contribute to the further innovation and application of nanocarriers derived from biological sources for the targeted treatment of breast cancer.
Collapse
Affiliation(s)
- Fahimeh Kazeminava
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Siamak Javanbakht
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Latifi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Mahmoud Abbaszadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Adibag
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Bisen AC, Sanap SN, Agrawal S, Biswas A, Mishra A, Verma SK, Singh V, Bhatta RS. Etiopathology, Epidemiology, Diagnosis, and Treatment of Fungal Keratitis. ACS Infect Dis 2024; 10:2356-2380. [PMID: 38847789 DOI: 10.1021/acsinfecdis.4c00203] [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: 07/13/2024]
Abstract
Fungal keratitis (FK) is a severe ocular condition resulting from corneal infection that is prevalent in tropical countries, particularly in developing regions of Asia and Africa. Factors like corneal lens misuse, inappropriate steroid use, and diagnostic challenges have provoked the epidemic. FK causes significant vision impairment, scarring, and ocular deformities. Accurate pathological diagnosis is crucial for effective therapeutic intervention. Topical antifungal therapy with surface healing medications proves effective in preventing fungal-borne ulcers. Managing FK requires a comprehensive understanding of fungal pathogenesis, guiding formulation strategies and preventive measures to curb global ocular blindness. This review provides in-depth insights into FK, covering etiology, epidemiology, pathogenesis, therapeutic interventions, antifungal resistance, limitations, prevention, and future perspectives on ocular surface disease management.
Collapse
Affiliation(s)
- Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
- Sophisticated Analytical Instrument Facility and Research, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sachin Nashik Sanap
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Arpon Biswas
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anjali Mishra
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sarvesh Kumar Verma
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Vaishali Singh
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
25
|
Yang L, An Y, Xu D, Dai F, Shao S, Lu Z, Liu G. Comprehensive Overview of Controlled Fabrication of Multifunctional Fluorescent Carbon Quantum Dots and Exploring Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309293. [PMID: 38342681 DOI: 10.1002/smll.202309293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/24/2024] [Indexed: 02/13/2024]
Abstract
In recent years, carbon dots (CDs) have garnered increasing attention due to their simple preparation methods, versatile performances, and wide-ranging applications. CDs can manifest various optical, physical, and chemical properties including quantum yield (QY), emission wavelength (Em), solid-state fluorescence (SSF), room-temperature phosphorescence (RTP), material-specific responsivity, pH sensitivity, anti-oxidation and oxidation, and biocompatibility. These properties can be effectively regulated through precise control of the CD preparation process, rendering them suitable for diverse applications. However, the lack of consideration given to the precise control of each feature of CDs during the preparation process poses a challenge in obtaining the requisite features for various applications. This paper is to analyze existing research and present novel concepts and ideas for creating CDs with different distinct features and applications. The synthesis methods of CDs are discussed in the first section, followed by a comprehensive overview of the important properties of CDs and the modification strategy. Subsequently, the application of CDs and their requisite properties are reviewed. Finally, the paper outlines the current challenges in controlling CDs properties and their applications, discusses potential solutions, and offers suggestions for future research.
Collapse
Affiliation(s)
- Lijuan Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yibo An
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Dazhuang Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Fan Dai
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Shillong Shao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zhixiang Lu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
26
|
Gautam S, Singh N, Marwaha D, Rai N, Sharma M, Tiwari P, Singh S, Kumar Bakshi A, Kumar A, Agarwal N, Prakash Shukla R, Ranjan Mishra P. Celastrol-loaded polymeric mixed micelles shows improved antitumor efficacy in 4 T1 bearing xenograft mouse model through spatial targeting. Int J Pharm 2024; 659:124234. [PMID: 38763310 DOI: 10.1016/j.ijpharm.2024.124234] [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: 12/09/2023] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
In this study, we have proposed a novel approach that combines hyaluronic acid (HA), folic acid (FA), and celastrol (CLS) within a polymeric micelle system (CLS-HF/MLs), offering a dual-action strategy against breast cancer. Polymeric mixed micelles were prepared through the thin-film hydration method, and comprehensive quality control parameters were established, encompassing particle size, polydispersity index, zeta potential, surface morphology, encapsulation efficiency, drug content, in vitro drug release, and storage stability assessment. The average particle size of CLS-HF/MLs micelles was found to be 120 nm and their drug loading and encapsulation efficiencies were 15.9 % and 89.52 %, respectively. The in vitro release data showed that the CLS-HF/MLs targeted mixed micelles displayed a prolonged release profile compared to the free drug. Additionally, the stability of the developed polymeric mixed micelles was maintained for up to 8 weeks of storage in terms of particle size and drug content. Furthermore, both flow cytometry and confocal laser scanning microscopy studies indicated a significant enhancement in the cellular uptake efficiency and cytotoxicity of CLS-HF/MLs mixed micelles against MCF-7 cell line. In terms of pharmacokinetic analysis, the half-life and AUC values of CLS-HF/MLs mixed micelles were found to be approximately 4.71- and 7.36-folds higher than the values of free drug (CLS), respectively. The CLS-HF/MLs micelles exhibited remarkable antitumor efficacy (almost complete ablation of the 4 T1-cell bearing tumor xenografts mouse model) due to the dual receptor (CD44 and folate) targeting effects with minimal side effects. When considering the cumulative findings of our present research, it becomes evident that mixed micelles designed for chemotherapy offer a promising and potentially effective therapeutic avenue for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Sanjay Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Ankit Kumar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
27
|
Bisen AC, Dubey A, Agrawal S, Biswas A, Rawat KS, Srivastava S, Bhatta RS. Recent updates on ocular disease management with ophthalmic ointments. Ther Deliv 2024; 15:463-480. [PMID: 38888757 DOI: 10.1080/20415990.2024.2346047] [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: 10/31/2023] [Accepted: 04/18/2024] [Indexed: 06/20/2024] Open
Abstract
Ophthalmic diseases can result in permanent vision loss and blindness. Convenient topical and systemic treatments are preferred to address these sight-threatening conditions. However, the unique anatomy of the eye presents challenges for drug delivery. Various ophthalmic ointment formulations have been developed to enhance bioavailability in the eye to prolong residence time and improve corneal permeability. This article explores a wide range of ocular diseases affecting individuals globally and how ointments are used to manage them. From eye to ocular barriers, this review focuses on published scientific research and formulation strategies for severe ocular complications using conventional topical ointments. Additionally, it delves through patented technologies and marketed formulations supporting the use of ointments in ocular drug delivery.
Collapse
Affiliation(s)
- Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
- Sophisticated Analytical Instrument Facility and Research, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Ayush Dubey
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- School of Pharmaceutical Sciences, CSJM University, Kanpur, 208024, Uttar Pradesh, India
| | - Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Arpon Biswas
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Kundan Singh Rawat
- Prof. Rajendra Singh Nanoscience & Chemistry D.S.B. Campus, Kumaun University, Nainital, 263001, Uttarakhand, India
| | - Saurabh Srivastava
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- School of Pharmaceutical Sciences, CSJM University, Kanpur, 208024, Uttar Pradesh, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| |
Collapse
|
28
|
Bisen AC, Mishra A, Agrawal S, Sanap SN, Biswas A, Verma SK, Bhatta RS. Codetermination of antimicrobial agents in rabbit tear fluid using LC-MS/MS assay: Insights into ocular pharmacokinetic study. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5031. [PMID: 38726684 DOI: 10.1002/jms.5031] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 08/24/2024]
Abstract
Managing ocular microbial infections typically requires pharmacotherapy using antibiotic eye drops, such as moxifloxacin hydrochloride (MFX), combined with an antifungal agent like amphotericin B (AB). We carried out and validated an LC-MS/MS assay to quantify these compounds in rabbit tear fluid in order to look into the pharmacokinetics of these two drugs. We employed a protein precipitation technique for the extraction of drugs under examination. A Waters Symmetry C18 column was used to separate the analytes and internal standard. The composition of the mobile phase was like (A) 0.1% v/v formic acid in water and (B) methanol. The detection of MFX and AB was accomplished through the utilization of positive ion electrospray ionization under multiple reaction monitoring mode. The linearity curves for both analytes exhibited an acceptable trendline across a concentration range of 2.34-300 ng/mL for MFX and 7.81-1000 ng/mL for AB in surrogate rabbit tear fluid. The lower limit of quantitation for MFX was 2.34 ng/mL, while for AB, it was 7.81 ng/mL. The approach was strictly validated, encompassing tests of selectivity, linearity (with r2 > 0.99), precision, accuracy, matrix effects, and stability. Consequently, we employed this method to evaluate the pharmacokinetics profiles of MFX and AB in rabbit tear fluid following single topical doses.
Collapse
Affiliation(s)
- Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Anjali Mishra
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Sachin Nashik Sanap
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Arpon Biswas
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Sarvesh Kumar Verma
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
29
|
Tiwari P, Yadav K, Shukla RP, Bakshi AK, Panwar D, Das S, Mishra PR. Extracellular vesicles-powered immunotherapy: Unleashing the potential for safer and more effective cancer treatment. Arch Biochem Biophys 2024; 756:110022. [PMID: 38697343 DOI: 10.1016/j.abb.2024.110022] [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: 03/20/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Cancer treatment has seen significant advancements with the introduction of Onco-immunotherapies (OIMTs). Although some of these therapies have received approval for use, others are either undergoing testing or are still in the early stages of development. Challenges persist in making immunotherapy widely applicable to cancer treatment. To maximize the benefits of immunotherapy and minimize potential side effects, it's essential to improve response rates across different immunotherapy methods. A promising development in this area is the use of extracellular vesicles (EVs) as novel delivery systems. These small vesicles can effectively deliver immunotherapies, enhancing their effectiveness and reducing harmful side effects. This article discusses the importance of integrating nanomedicines into OIMTs, highlighting the challenges with current anti-OIMT methods. It also explores key considerations for designing nanomedicines tailored for OIMTs, aiming to improve upon existing immunotherapy techniques. Additionally, the article looks into innovative approaches like biomimicry and the use of natural biomaterial-based nanocarriers (NCs). These advancements have the potential to transform the delivery of immunotherapy. Lastly, the article addresses the challenges of moving OIMTs from theory to clinical practice, providing insights into the future of using advanced nanotechnology in cancer treatment.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Sweety Das
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
30
|
Yadav K, Vijayalakshmi R, Kumar Sahu K, Sure P, Chahal K, Yadav R, Sucheta, Dubey A, Jha M, Pradhan M. Exosome-Based Macromolecular neurotherapeutic drug delivery approaches in overcoming the Blood-Brain barrier for treating brain disorders. Eur J Pharm Biopharm 2024; 199:114298. [PMID: 38642716 DOI: 10.1016/j.ejpb.2024.114298] [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: 02/03/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Delivering drugs to the brain is a complex challenge in medical research, particularly for disorders like Alzheimer's and Parkinson's. The blood-brain barrier restricts the entry of many therapeutic molecules, hindering their effectiveness. Nanoparticles, a potential solution, face issues like toxicity and limited approvals. A new avenue explores the use of small extracellular vesicles (sEVs), i.e., exosomes, as natural carriers for drug delivery. sEVs, tiny structures below 150 nm, show promise due to their minimal immune response and ability to precisely deliver drugs. This review focuses on the potential of sEVs-based drug delivery systems for treating neurological disorders, brain cancers, and other brain-related issues. Notably, bioengineered sEVs-carrying therapeutic compounds exhibit promise in early studies. The unique features of sEVs, such as their small size and natural properties, position them as candidates to overcome challenges in drug delivery to the brain. Ongoing clinical trials and research into sEVs behavior within the body further highlight their potential for revolutionizing drug delivery and addressing complex brain conditions.
Collapse
Affiliation(s)
- Krishna Yadav
- Raipur Institute of Pharmaceutical Education and Research, Sarona, Raipur, Chhattisgarh 492010, India
| | - R Vijayalakshmi
- Department of Pharmaceutical Analysis, GIET School of Pharmacy, Chaitanya Knowledge City, Rajahmundry, AP, 533296, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Pavani Sure
- Department of Pharmaceutics, Vignan Institute of Pharmaceutical Sciences, Hyderabad, Telangana, India
| | - Kavita Chahal
- Department of Botany, Government Model Science College Jabalpur, Madhya Pradesh, India
| | - Renu Yadav
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana, 122103, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Sohna Road, Gurugram, Haryana, 122103, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru-575018, Karnataka, India
| | - Megha Jha
- Department of Life Science, Mansarovar Global University, Sehore, M.P., India
| | - Madhulika Pradhan
- Gracious College of Pharmacy, Abhanpur, Chhattisgarh, 493661, India.
| |
Collapse
|
31
|
Omidian H, Wilson RL, Cubeddu LX. Quantum Dot Research in Breast Cancer: Challenges and Prospects. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2152. [PMID: 38730959 PMCID: PMC11085412 DOI: 10.3390/ma17092152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024]
Abstract
The multifaceted role of quantum dots (QDs) in breast cancer research highlights significant advancements in diagnostics, targeted therapy, and drug delivery systems. This comprehensive review addresses the development of precise imaging techniques for early cancer detection and the use of QDs in enhancing the specificity of therapeutic delivery, particularly in challenging cases like triple-negative breast cancer (TNBC). The paper also discusses the critical understanding of QDs' interactions with cancer cells, offering insights into their potential for inducing cytotoxic effects and facilitating gene therapy. Limitations such as biocompatibility, toxicity concerns, and the transition from laboratory to clinical practice are critically analyzed. Future directions emphasize safer, non-toxic QD development, improved targeting mechanisms, and the integration of QDs into personalized medicine, aiming to overcome the current challenges and enhance breast cancer management.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (R.L.W.); (L.X.C.)
| | | | | |
Collapse
|
32
|
Tiwari P, Shukla RP, Yadav K, Panwar D, Agarwal N, Kumar A, Singh N, Bakshi AK, Marwaha D, Gautam S, Rai N, Mishra PR. Exploring nanocarriers as innovative materials for advanced drug delivery strategies in onco-immunotherapies. J Mol Graph Model 2024; 128:108702. [PMID: 38219505 DOI: 10.1016/j.jmgm.2024.108702] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/16/2023] [Accepted: 01/02/2024] [Indexed: 01/16/2024]
Abstract
In recent years, Onco-immunotherapies (OIMTs) have been shown to be a potential therapy option for cancer. Several immunotherapies have received regulatory approval, while many others are now undergoing clinical testing or are in the early stages of development. Despite this progress, a large number of challenges to the broad use of immunotherapies to treat cancer persists. To make immunotherapy more useful as a treatment while reducing its potentially harmful side effects, we need to know more about how to improve response rates to different types of immunotherapies. Nanocarriers (NCs) have the potential to harness immunotherapies efficiently, enhance the efficiency of these treatments, and reduce the severe adverse reactions that are associated with them. This article discusses the necessity to incorporate nanomedicines in OIMTs and the challenges we confront with current anti-OIMT approaches. In addition, it examines the most important considerations for building nanomedicines for OIMT, which may improve upon current immunotherapy methods. Finally, it highlights the applications and future scenarios of using nanotechnology.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Ankit Kumar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
33
|
Mugundhan SL, Mohan M. Nanoscale strides: exploring innovative therapies for breast cancer treatment. RSC Adv 2024; 14:14017-14040. [PMID: 38686289 PMCID: PMC11056947 DOI: 10.1039/d4ra02639j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
Breast cancer (BC) is a predominant malignancy in women that constitutes approximately 30% of all cancer cases and has a mortality rate of 14% in recent years. The prevailing therapies include surgery, chemotherapy, and radiotherapy, each with its own limitations and challenges. Despite oral or intravenous administration, there are numerous barriers to accessing anti-BC agents before they reach the tumor site, including physical, physiological, and biophysical barriers. The complexity of BC pathogenesis, attributed to a combination of endogenous, chronic, intrinsic, extrinsic and genetic factors, further complicates its management. Due to the limitations of existing cancer treatment approaches, there is a need to explore novel, efficacious solutions. Nanodrug delivery has emerged as a promising avenue in cancer chemotherapy, aiming to enhance drug bioavailability while mitigating adverse effects. In contrast to conventional chemotherapy, cancer nanotechnology leverages improved permeability to achieve comprehensive disruption of cancer cells. This approach also presented superior pharmacokinetic profiles. The application of nanotechnology in cancer therapeutics includes nanotechnological tools, but a comprehensive review cannot cover all facets. Thus, this review concentrates specifically on BC treatment. The focus lies in the successful implementation of systematic nanotherapeutic strategies, demonstrating their superiority over conventional methods in delivering anti-BC agents. Nanotechnology-driven drug delivery holds immense potential in treating BC. By surmounting multiple barriers and capitalizing on improved permeability, nanodrug delivery has demonstrated enhanced efficacy and reduced adverse effects compared to conventional therapies. This review highlights the significance of systematic nanotherapy approaches, emphasizing the evolving landscape of BC management.
Collapse
Affiliation(s)
- Sruthi Laakshmi Mugundhan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology SRM Nagar Kattankulathur 603203 Tamil Nadu India
| | - Mothilal Mohan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology SRM Nagar Kattankulathur 603203 Tamil Nadu India
| |
Collapse
|
34
|
Cao J, Lv G, Wei F. Engineering exosomes to reshape the immune microenvironment in breast cancer: Molecular insights and therapeutic opportunities. Clin Transl Med 2024; 14:e1645. [PMID: 38572668 PMCID: PMC10993163 DOI: 10.1002/ctm2.1645] [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: 11/28/2023] [Revised: 02/19/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Breast cancer remains a global health challenge, necessitating innovative therapeutic approaches. Immunomodulation and immunotherapy have emerged as promising strategies for breast cancer treatment. Engineered exosomes are the sort of exosomes modified with surface decoration and internal therapeutic molecules. Through suitable modifications, engineered exosomes exhibit the capability to overcome the limitations associated with traditional therapeutic approaches. This ability opens up novel avenues for the development of more effective, personalized, and minimally invasive interventions. MAIN BODY In this comprehensive review, we explore the molecular insights and therapeutic potential of engineered exosomes in breast cancer. We discuss the strategies employed for exosome engineering and delve into their molecular mechanisms in reshaping the immune microenvironment of breast cancer. CONCLUSIONS By elucidating the contribution of engineered exosomes to breast cancer immunomodulation, this review underscores the transformative potential of this emerging field for improving breast cancer therapy. HIGHLIGHTS Surface modification of exosomes can improve the targeting specificity. The engineered exosome-loaded immunomodulatory cargo regulates the tumour immune microenvironment. Engineered exosomes are involved in the immune regulation of breast cancer.
Collapse
Affiliation(s)
- Jilong Cao
- Party Affairs and Administration Officethe Fourth Affiliated Hospital of China Medical UniversityShenyangP. R. China
| | - Gang Lv
- Department of Thyroid and Breast SurgeryChaohu Hospital of Anhui Medical UniversityChaohuP. R. China
| | - Fang Wei
- Department of General Surgerythe Fourth Affiliated Hospital of China Medical UniversityShenyangP. R. China
| |
Collapse
|
35
|
Yin W, Ma H, Qu Y, Wang S, Zhao R, Yang Y, Guo ZN. Targeted exosome-based nanoplatform for new-generation therapeutic strategies. Biomed Mater 2024; 19:032002. [PMID: 38471163 DOI: 10.1088/1748-605x/ad3310] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [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
Exosomes, typically 30-150 nm in size, are lipid-bilayered small-membrane vesicles originating in endosomes. Exosome biogenesis is regulated by the coordination of various mechanisms whereby different cargoes (e.g. proteins, nucleic acids, and lipids) are sorted into exosomes. These components endow exosomes with bioregulatory functions related to signal transmission and intercellular communication. Exosomes exhibit substantial potential as drug-delivery nanoplatforms owing to their excellent biocompatibility and low immunogenicity. Proteins, miRNA, siRNA, mRNA, and drugs have been successfully loaded into exosomes, and these exosome-based delivery systems show satisfactory therapeutic effects in different disease models. To enable targeted drug delivery, genetic engineering and chemical modification of the lipid bilayer of exosomes are performed. Stimuli-responsive delivery nanoplatforms designed with appropriate modifications based on various stimuli allow precise control of on-demand drug delivery and can be utilized in clinical treatment. In this review, we summarize the general properties, isolation methods, characterization, biological functions, and the potential role of exosomes in therapeutic delivery systems. Moreover, the effective combination of the intrinsic advantages of exosomes and advanced bioengineering, materials science, and clinical translational technologies are required to accelerate the development of exosome-based delivery nanoplatforms.
Collapse
Affiliation(s)
- Wenjing Yin
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Hongyin Ma
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Yang Qu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Siji Wang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Ruoyu Zhao
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
- Neuroscience Research Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| |
Collapse
|
36
|
Fernández-Borbolla A, García-Hevia L, Fanarraga ML. Cell Membrane-Coated Nanoparticles for Precision Medicine: A Comprehensive Review of Coating Techniques for Tissue-Specific Therapeutics. Int J Mol Sci 2024; 25:2071. [PMID: 38396747 PMCID: PMC10889273 DOI: 10.3390/ijms25042071] [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: 12/06/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Nanoencapsulation has become a recent advancement in drug delivery, enhancing stability, bioavailability, and enabling controlled, targeted substance delivery to specific cells or tissues. However, traditional nanoparticle delivery faces challenges such as a short circulation time and immune recognition. To tackle these issues, cell membrane-coated nanoparticles have been suggested as a practical alternative. The production process involves three main stages: cell lysis and membrane fragmentation, membrane isolation, and nanoparticle coating. Cell membranes are typically fragmented using hypotonic lysis with homogenization or sonication. Subsequent membrane fragments are isolated through multiple centrifugation steps. Coating nanoparticles can be achieved through extrusion, sonication, or a combination of both methods. Notably, this analysis reveals the absence of a universally applicable method for nanoparticle coating, as the three stages differ significantly in their procedures. This review explores current developments and approaches to cell membrane-coated nanoparticles, highlighting their potential as an effective alternative for targeted drug delivery and various therapeutic applications.
Collapse
Affiliation(s)
- Andrés Fernández-Borbolla
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| | - Lorena García-Hevia
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| | - Mónica L. Fanarraga
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| |
Collapse
|
37
|
Bisen AC, Bhatta RS. Ocular bioanalysis of moxifloxacin and ketorolac tromethamine in rabbit lacrimal matrix using liquid chromatography-tandem mass spectrometry. Bioanalysis 2024; 16:219-232. [PMID: 38197429 DOI: 10.4155/bio-2023-0233] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Abstract
Aim: The fixed-dose combination of moxifloxacin (MOXI) and ketorolac tromethamine (KTR) is widely used for the treatment of bacterial keratitis. Thus, a new LC-MS/MS method was developed to determine MOXI and KTR in lacrimal fluid. Methods: Bioanalysis was performed using a Shimadzu 8050 LC-MS/MS in electrospray ionization-positive mode and the method was validated per US FDA guidelines. Isocratic separation was performed with a Waters Symmetry C18 column using methanol and 0.1% formic acid containing deionized water (85:15, v/v). Results & conclusion: An easy, quick and selective method was established and applied to assess the ocular pharmacokinetic profile of a commercially available formulation containing MOXI and KTR. Based on the pharmacokinetic data, this work describes pharmacokinetics-based dosage regimen calculations and their clinical significance.
Collapse
Affiliation(s)
- Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad, Uttar Pradesh, 201002, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|