1
|
Khoshdooz S, Khoshdooz P, Bonyad R, Bonyad A, Sheidaei S, Nosrati R. Cubosomes-based hydrogels; A promising advancement for drug delivery. Int J Pharm 2025; 674:125510. [PMID: 40132766 DOI: 10.1016/j.ijpharm.2025.125510] [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: 12/29/2024] [Revised: 03/01/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Hydrogels have so far shown promising opportunities for possible drug delivery applications. Cubosomes (Cub), bicontinuous cubic phase liquid crystals, possess several characteristics that make them appealing as a versatile medium for drug administration. They have been regarded as prospective nanocarriers for drugs, offering a promising alternative to liposomes as a drug delivery method. Cub have the ability to encapsulate lipophilic, hydrophilic, and amphiphilic medicines. Hydrogels have recently shown significant interest in using Cub-based formulations. This paper examines the current advancements in biodegradable Cub-based hydrogels (Cubogel) for intelligent medication delivery to various organs. In conclusion, this paper briefly discusses the prospects and problems of hydrogels based on Cub.
Collapse
Affiliation(s)
- Sara Khoshdooz
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parisa Khoshdooz
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reihaneh Bonyad
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Bonyad
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sina Sheidaei
- Department of Chemistry, Faculty of Science, University of Guilan, Iran
| | - Rahim Nosrati
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
2
|
Luo F, Deng Y, Angelov B, Angelova A. Melatonin and the nervous system: nanomedicine perspectives. Biomater Sci 2025. [PMID: 40231558 DOI: 10.1039/d4bm01609b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
The mechanism of action of melatonin on the nervous system, sleep, cognitive deficits, and aging is not fully understood. Neurodegenerative diseases (ND) are one of the leading causes of disability and mortality worldwide. Sleeping and cognitive impairments also represent common and serious public health problems, particularly deteriorating with the aging process. Melatonin, as a neuromodulatory hormone, regulates circadian rhythms and the sleep-wake cycle, with functions extending to antioxidant, anti-inflammatory, neuroprotective, and anti-aging properties. However, melatonin is a hydrophobic compound with relatively low water solubility and a short half-life. While melatonin can cross the blood-brain barrier, exogenous melatonin administered orally or intravenously has poor bioavailability, undergoes rapid metabolism in the circulation, and shows limited brain accumulation, ultimately compromising its therapeutic efficacy. In recent years, the convergence of melatonin research with nanomedicine ensures safe therapeutic uses, limited drug degradation, and perspectives for targeted drug delivery to the central nervous system. Here we outline the promising neurotherapeutic properties of nanomaterials as carriers loaded with melatonin drug alone or in combinations with other active molecules.
Collapse
Affiliation(s)
- Fucen Luo
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, F-91400 Orsay, France.
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, No.1, Jinlian Road, Longwan District, Wenzhou, Zhejiang 325001, China
| | - Borislav Angelov
- Extreme Light Infrastructure ERIC, Department of Structural Dynamics, CZ-25241 Dolni Brezany, Czech Republic
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, F-91400 Orsay, France.
| |
Collapse
|
3
|
Boulbazine M, Djellala I, Boudjahem AG. A density functional theory study on the adsorption of the β-lapachone anti-cancer drug onto the MB 11N 12 (M = au, Rh and Ru) nanoclusters as a drug delivery. J Mol Graph Model 2025; 138:109044. [PMID: 40233483 DOI: 10.1016/j.jmgm.2025.109044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/22/2025] [Accepted: 03/30/2025] [Indexed: 04/17/2025]
Abstract
The structural and electronic properties of the pristine and metal(M)-doped B12N12 (M = Ru, Rh and Au) nanoclusters were systematically analyzed using DFT calculations. The results indicate that the B12N12 behaves like a semiconductor with a substantial HOMO-LUMO energy gap of 6.75 eV. The introduction of the metal dopants (Ru, Rh and Au) in the pristine leads to a significant reduction of its gap energy with a variation in Eg ranging from 48.7 % to 80 %. This substantial decrease in the value of Eg underlines the crucial role that the metal can play in the electronic structure and the catalytic performance of the resulting material. The performance of the B12N12 cluster has been greatly improved with doping, and the doped clusters can be used in advanced technological applications. In order to explore the surface reactivity and sensing performance of the B12N12 nanocluster and their counterparts doped with transition metals such as Ru, Rh and Au towards the molecule cancer drugs, we systematically studied the adsorption behavior of the β-lapachone drug onto their surface. The molecule drug exhibited strong binding to B12N12 with adsorption energies of - 31.42 to - 40.0 kcal mol-1 for the two most stable configurations. For the metal-doped B12N12 nanoclusters, the highest adsorption energy (- 68.0 kcal mol-1) was obtained for the cluster doped by the Ru atom. The charge transfer analysis confirmed that β-lapachone gives electrons to nanoclusters, improving their chemical stability. In addition, the evaluation of the solvation energies indicates an improvement in drug delivery performance in biological environment. This study demonstrates the promise of the metal-doped B12N12 nanoclusters as effective carriers for the β-lapachone drug, highlighting their stability, reactivity and suitability for drug delivery applications.
Collapse
Affiliation(s)
- Mouhssin Boulbazine
- The Division of Research in School and Its Environment, National Institute for Research in Education, BP 193, Industrial Zone, Oued Romane-El Achour, Algeria.
| | - Imane Djellala
- Laboratory of Computational Chemistry and Nanostructures, Department of Material Sciences, University of 8 Mai 1945, Guelma, Algeria.
| | - Abdel-Ghani Boudjahem
- Computational Catalysis Group, Laboratory of Applied Chemistry, University of 8 Mai 1945, Guelma, Algeria.
| |
Collapse
|
4
|
Ultimo A, Jain A, Gomez-Gonzalez E, Alex TS, Moreno-Borrallo A, Jana S, Ghosh S, Ruiz-Hernandez E. Nanotherapeutic Formulations for the Delivery of Cancer Antiangiogenics. Mol Pharm 2025. [PMID: 40184281 DOI: 10.1021/acs.molpharmaceut.4c00822] [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: 04/06/2025]
Abstract
Antiangiogenic medications for cancer treatment have generally failed in showing substantial benefits in terms of prolonging life on their own; their effects are noticeable only when combined with chemotherapy. Moreover, treatments based on prolonged antiangiogenics administration have demonstrated to be ineffective in stopping tumor progression. In this scenario, nanotherapeutics can address certain issues linked to existing antiangiogenic treatments. More specifically, they can provide the ability to target the tumor's blood vessels to enhance drug accumulation and manage release, ultimately decreasing undesired side effects. Additionally, they enable the administration of multiple angiogenesis inhibitors at the same time as chemotherapy. Key reports in this field include the design of polymeric nanoparticles, inorganic nanoparticles, vesicles, and hydrogels for loading antiangiogenic substances like endostatin and interleukin-12. Furthermore, nanoformulations have been proposed to efficiently control relevant pro-angiogenic pathways such as VEGF, Tie2/Angiopoietin-1, HIF-1α/HIF-2α, and TGF-β, providing powerful approaches to block tumor growth and metastasis. In this article, we outline a selection of nanoformulations for antiangiogenic treatments for cancer that have been developed in the past ten years.
Collapse
Affiliation(s)
- Amelia Ultimo
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Ayushi Jain
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Elisabet Gomez-Gonzalez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Thomson Santosh Alex
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Almudena Moreno-Borrallo
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Sukanya Jana
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Shubhrima Ghosh
- Trinity Translational Medicine Institute, Trinity College Dublin, the University of Dublin, St. James's Hospital, Dublin 8 D08 NHY1, Ireland
- School of Biological, Health and Sports Sciences, Technological University Dublin, Grangegorman Lower, Dublin 7 D07 ADY7, Ireland
| | - Eduardo Ruiz-Hernandez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| |
Collapse
|
5
|
Zhang H, Chen H, Guo G, Lin J, Chen X, Huang P, Lin C, Lin H, Lu Y, Lin J, Li X, Zhang W. Nanotechnology in prostate cancer: a bibliometric analysis from 2004 to 2023. Discov Oncol 2025; 16:451. [PMID: 40175778 PMCID: PMC11965044 DOI: 10.1007/s12672-025-02265-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/28/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Prostate cancer (PC) contributes to male mortality worldwide. The objective of this study is to comprehensively depict the scientific accomplishments and research trends in nanotechnology for PC applications. METHODS Utilizing the Web of Science Core Collection database, publications were gathered on the basis of inclusion and selection criteria. The publications were analyzed and visualized using VOSviewer, R-studio and CiteSpace software tools. RESULTS A total of 1949 studies were incorporated. Farokhzad was the most productive author. The United States and China released 58.13% of the total publications. The Chinese Academy of Sciences was the most influential institution, and the International Journal of Nanomedicine stood out as a prominent journal in this field. The most frequently referenced publication and research subject category were identified. The most extensively investigated area was nanoparticle-based drug delivery, while recent research has focused on anticancer with novel nanocarriers. CONCLUSION A bibliometric analysis in the PC and nanotechnology was conducted between 2004 and 2023. The overview and characteristics of the publications were identified. We discussed the application and restrictions faced by nanotechnology in PC management. The study of nanotechnology in PC treatment needs to be further studied.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Hongpeng Chen
- Department of Oncology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Gaowei Guo
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Jinming Lin
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Xiaosheng Chen
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Peidong Huang
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Chuqi Lin
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Huirong Lin
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Yong Lu
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Jieming Lin
- Department of Operating Room, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Xinji Li
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China.
| | - Wei Zhang
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China.
| |
Collapse
|
6
|
Xu G, Zhang Q, Cheng R, Qu J, Li W. Survival strategies of cancer cells: the role of macropinocytosis in nutrient acquisition, metabolic reprogramming, and therapeutic targeting. Autophagy 2025; 21:693-718. [PMID: 39817564 PMCID: PMC11925119 DOI: 10.1080/15548627.2025.2452149] [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: 10/09/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Macropinocytosis is a nonselective form of endocytosis that allows cancer cells to largely take up the extracellular fluid and its contents, including nutrients, growth factors, etc. We first elaborate meticulously on the process of macropinocytosis. Only by thoroughly understanding this entire process can we devise targeted strategies against it. We then focus on the central role of the MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) in regulating macropinocytosis, highlighting its significance as a key signaling hub where various pathways converge to control nutrient uptake and metabolic processes. The article covers a comprehensive analysis of the literature on the molecular mechanisms governing macropinocytosis, including the initiation, maturation, and recycling of macropinosomes, with an emphasis on how these processes are hijacked by cancer cells to sustain their growth. Key discussions include the potential therapeutic strategies targeting macropinocytosis, such as enhancing drug delivery via this pathway, inhibiting macropinocytosis to starve cancer cells, blocking the degradation and recycling of macropinosomes, and inducing methuosis - a form of cell death triggered by excessive macropinocytosis. Targeting macropinocytosis represents a novel and innovative approach that could significantly advance the treatment of cancers that rely on this pathway for survival. Through continuous research and innovation, we look forward to developing more effective and safer anti-cancer therapies that will bring new hope to patients.Abbreviation: AMPK: AMP-activated protein kinase; ASOs: antisense oligonucleotides; CAD: carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase; DC: dendritic cell; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; ERBB2: erb-b2 receptor tyrosine kinase 2; ESCRT: endosomal sorting complex required for transport; GAP: GTPase-activating protein; GEF: guanine nucleotide exchange factor; GRB2: growth factor receptor bound protein 2; LPP: lipopolyplex; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; NSCLC: non-small cell lung cancer; PADC: pancreatic ductal adenocarcinoma; PDPK1: 3-phosphoinositide dependent protein kinase 1; PI3K: phosphoinositide 3-kinase; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns(3,4,5)P3: phosphatidylinositol-(3,4,5)-trisphosphate; PtdIns(4,5)P2: phosphatidylinositol-(4,5)-bisphosphate; PTT: photothermal therapies; RAC1: Rac family small GTPase 1; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; RTKs: receptor tyrosine kinases; SREBF: sterol regulatory element binding transcription factor; TFEB: transcription factor EB; TNBC: triple-negative breast cancer; TSC2: TSC complex subunit 2; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Guoshuai Xu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Qinghong Zhang
- Emergency Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Renjia Cheng
- Department of Intensive Care Medicine, The General Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang, Liaoning, China
| | - Jun Qu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Wenqiang Li
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
7
|
Kumar P, Ashique S, Sharma H, Yasmin S, Islam A, Mandal S, Gowda BHJ, Khalid M, Ansari MY, Singh M, Ehsan I, Taj T, Taghizadeh-Hesary F. A narrative review on the use of Green synthesized metallic nanoparticles for targeted cancer therapy. Bioorg Chem 2025; 157:108305. [PMID: 40022847 DOI: 10.1016/j.bioorg.2025.108305] [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: 12/23/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Cancer is a leading cause of death worldwide. While traditional and synthetic medical therapies are in place for cancer treatment, their effectiveness is hindered by various limitations, such as toxic side effects, limited availability, and high costs. In recent years, a promising alternative approach has emerged in the form of green-synthesized metallic nanoparticles (MNPs), which offer targeted cancer therapy. These nanoparticles (NPs) have garnered significant attention from cancer researchers owing to their natural or surface-induced anticancer properties, versatility of metals as agents, and eco-friendly nature. This approach may positively impact healthy cells surrounding the cancerous cells. Green-synthesized MNPs have gained popularity in cancer management because of their ease of handling in the laboratory and the affordability of starting materials compared to synthetic methods. This review analyzes green-synthesized MNPs for targeted cancer therapy, highlighting tumor-targeting strategies, synthesis methods, and clinical challenges. Unlike general reviews, it compares plant-, microbial-, and enzyme-mediated synthesis approaches, emphasizing their impact on nanoparticle stability, functionalization, and interactions with the tumor microenvironment for enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Prashant Kumar
- SRM Modinagar College of Pharmacy, SRMIST Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, 201204, India
| | - Sumel Ashique
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, West Bengal 711316, India.
| | - Himanshu Sharma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, (UP), India
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow 226026, Uttar Pradesh, India
| | - Subhajit Mandal
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Mohammad Khalid
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Asir-Abha 61421, Saudi Arabia
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India; Ibne Seena College of Pharmacy, Azmi Vidya Nagri Anjhi Shahabad, Hardoi-241124 Uttar Pradesh (U.P.) India.
| | - Mansi Singh
- Research Scholar, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Iman Ehsan
- School of Pharmacy Sister Nivedita University, Kolkata-700156, WB, India
| | - Tahreen Taj
- Research Scholar, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India; Department of Pharmacology, Yenepoya Pharmacy college and research centre, Yenepoya (Deemed to be) university, Mangalore 575018, India
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Clinical Oncology, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Lv Y, Jiang M, Ouyang Y, Zheng X, Zhang L, Yao J, Hu L, Zhao J, Li Z, Wang S. Sodium Butyrate-Loaded Microspheres With Enhanced Bioavailability for Targeted Treatment of Intestinal Barrier Injury. Adv Healthc Mater 2025; 14:e2402773. [PMID: 39629540 DOI: 10.1002/adhm.202402773] [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: 07/26/2024] [Revised: 11/09/2024] [Indexed: 01/16/2025]
Abstract
Intestinal barrier dysfunction is related to diseases such as inflammatory bowel disease (IBD) and severe acute pancreatitis (SAP). Butyrate and its derivatives (e.g., sodium butyrate (SB)) can alleviate gut inflammatory responses. Nevertheless, these substances usually cannot fully exert protective effects due to low bioavailability. This research aimed to offer microspheres for treating intestinal barrier injury. Sodium alginate solution is prepared to dissolve SB, followed by mixing with chitosan (CS)-protocatechuic aldehyde (PA)/calcium chloride solution. The required CS-PA/calcium alginate/SB (CPC/SB) microspheres are formed in this manner. Subsequently, the therapeutic effects of CPC/SB microspheres on intestinal barrier injury through in vivo dextran sulfate sodium salt (DSS)-induced IBD and sodium taurocholate (STC)-induced SAP models is explored. Results: The CPC/SB microspheres exhibited excellent antioxidant properties. In vivo bioluminescence imaging experiment confirmed the microspheres effectively targeted the inflammatory gut in IBD. Further in vivo experimental results indicated the microspheres significantly repaired intestinal barrier damage, exerting protective effects in IBD and SAP. Additionally, 16S rDNA sequencing explained the microspheres can regulate the balance between harmful and beneficial bacteria (such as Alistipes, Odoribacter, and Rikenellaceae RC9). This study provides a possible synthetic strategy of microsphere carriers to serve as a potential therapeutic tool for intestinal barrier injury.
Collapse
Affiliation(s)
- Yanwei Lv
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Mengni Jiang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Yongliang Ouyang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai, 200093, China
| | - Xiaoyi Zheng
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Liang Zhang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai, 200093, China
| | - Jinpeng Yao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Lianghao Hu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Jiulong Zhao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai, 200093, China
| |
Collapse
|
9
|
Selim S, Almuhayawi MS, Alruhaili MH, Saddiq AA, Baghdadi AM, Atta RMS, Al Jaouni SK. Synthesis, characterization, anticancer, antibacterial and antifungal activities of nanocomposite based on tertiary metal oxide Fe 2O 3@CuO@ZnONPs, starch, ethylcellulose and collagen. Int J Biol Macromol 2025; 301:140376. [PMID: 39880259 DOI: 10.1016/j.ijbiomac.2025.140376] [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: 10/29/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
This study aimed to synthesize a nanocomposite based on tertiary metal oxide Fe2O3@CuO@ZnONPs, starch, ethylcellulose, and collagen, as well as evaluate its biological activities. The prepared nanocomposites were characterized using physicochemical analysis, which included FTIR, XRD, and DLS. Additionally, topographical analysis using FI-SEM, EDX, mapping, HR-TEM, and SAED affirmed the molecular structure and nanosized of formulated nanocomposites. Moreover, DLS performed a size of free nanocomposite (Bnanocomp) and trimetallic loaded nanocomposite (Lnanocomp) as 158 and 105 nm, respectively. The synthesized loaded nanocomposite with metal oxides (Lnanocomp) was assessed for cytotoxicity on normal and cancerous cell lines. Results revealed that the IC50 of Lnanocomp toward Wi-38 normal cell line was 196.4 μg/mL; this confirms that Lnanocomp is non-toxic and safe in use. Moreover, Lnanocomp displayed anticancer activity against Hep-G2 with IC50 53.7 μg/mL. Furthermore, Lnanocomp displayed potential antibacterial activity toward E. coli, P. aeruginosa, S. typhimurium, S. aureus, and B. subtilis with MICs 50, 50, 12.5, 50, and 25 μg/mL, respectively. Also, Lnanocomp exhibited antifungal activity where MIC was 200, 50, and 100 μg/mL toward C. albicans, A. fumigatus, and A. brasilienisis respectively. In conclusion, the prepared Fe2O3@CuO@ZnONPs-based nanocomposite shows promising synergetic antibacterial, antifungal, and anticancer activities with state biocompatibility.
Collapse
Affiliation(s)
- Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia.
| | - Mohammed S Almuhayawi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Mohammed H Alruhaili
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Special Infectious Agents Unit, King Fahad Medical Research Center, King AbdulAziz University, Jeddah, Saudi Arabia.
| | - Amna A Saddiq
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia.
| | - Afra Mohammed Baghdadi
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia.
| | - Roba M S Atta
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia.
| | - Soad K Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
10
|
Biswas R, Mondal S, Ansari MA, Sarkar T, Condiuc IP, Trifas G, Atanase LI. Chitosan and Its Derivatives as Nanocarriers for Drug Delivery. Molecules 2025; 30:1297. [PMID: 40142072 PMCID: PMC11946192 DOI: 10.3390/molecules30061297] [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: 02/04/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Chitosan (CS) occurs naturally as an alkaline polysaccharide and has been demonstrated to have several activities of a biological nature. Additionally, as CS chains have functional hydroxyl and amino groups that are active, their applications can be expanded by chemically or molecularly altering the molecules to incorporate new functional groups. Due to its outstanding qualities, including biodegradability, biocompatibility, non-toxicity, and accessibility, it has received significant interest in all areas of biomedicine and nanomaterials being extremely promising as drug nanocarrier. The last decades have produced a lot of interest in CS-based nanoparticles (CSNPs), with an increasing number of research papers from around 1500 in 2015 to almost 5000 in 2024. The degree of crosslinking, the particulate system's shape, size, and density, in addition to the drug's physical and chemical properties, all have a role in how the drug is transported and released from CSNPs. When creating potential drug delivery systems based on CSNPs, all these factors must be considered. In earlier, CSNPs were employed to enhance the pharmacotherapeutics, pharmacokinetics, and solubility properties of drugs. By investigating its positively charged characteristics and changeable functional groups, CS has evolved into a versatile drug delivery system. The drug release from CSNPs will definitely be influenced by various changes to the functional groups, charges, and polymer backbone. This review mainly discusses the most important results published in the last decade. Despite the promising advantages of CSNPs, challenges related to the translation into clinical stages remain and further in vitro and in vivo studies are mandatory.
Collapse
Affiliation(s)
- Ranu Biswas
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, WB, India; (R.B.); (S.M.); (M.A.A.); (T.S.)
| | - Sourav Mondal
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, WB, India; (R.B.); (S.M.); (M.A.A.); (T.S.)
| | - Md Ahesan Ansari
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, WB, India; (R.B.); (S.M.); (M.A.A.); (T.S.)
| | - Tanima Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, WB, India; (R.B.); (S.M.); (M.A.A.); (T.S.)
| | - Iustina Petra Condiuc
- Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Gisela Trifas
- “Cristofor Simionescu” Faculty of Chemical Engineering and Environmental Protection, “Gheorghe Asachi” Technical University of Iasi, 700050 Iasi, Romania;
| | - Leonard Ionut Atanase
- “Cristofor Simionescu” Faculty of Chemical Engineering and Environmental Protection, “Gheorghe Asachi” Technical University of Iasi, 700050 Iasi, Romania;
- Faculty of Medicine, “Apollonia” University of Iasi, 700511 Iasi, Romania
- Academy of Romanian Scientists, 050045 Bucharest, Romania
| |
Collapse
|
11
|
Pareek A, Kumar D, Pareek A, Gupta MM. Advancing Cancer Therapy with Quantum Dots and Other Nanostructures: A Review of Drug Delivery Innovations, Applications, and Challenges. Cancers (Basel) 2025; 17:878. [PMID: 40075725 PMCID: PMC11898779 DOI: 10.3390/cancers17050878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025] Open
Abstract
Nanotechnology, particularly quantum dots (QDs), has ushered in a transformative era in the pharmaceutical and medical industries, offering notable opportunities for nanoscale advancements. These nanoscale particles, known for their exceptional optical properties and quantum confinement, have emerged as indispensable tools in cancer drug delivery and bioimaging. This review delves into various drug conjugation techniques with QDs, including covalent linking, non-covalent conjugation, click chemistry, disulfide linkage, and pH-sensitive linkage. Each method provides distinct advantages, such as enhanced stability, reversibility, specificity, and controlled drug release. Moreover, QDs have demonstrated significant promise in oncology by efficiently delivering drugs to cancerous tissues while minimising systemic toxicity. Investigations into their applications in different cancers, such as blood, brain, cervical, breast cancers, etc., reveal their efficacy in targeted drug delivery, real-time imaging, and improved therapeutic outcomes. However, challenges such as potential toxicity, stability, pharmacokinetics, and targeting specificity must be addressed to fully harness the benefits of QDs in cancer therapy. Future research should focus on developing biocompatible QDs, optimising conjugation techniques, and elucidating their safety profiles and long-term effects in biological systems. Overall, QDs represent a promising frontier in cancer treatment, offering multifaceted capabilities that hold the potential for enhanced therapeutic outcomes and reduced side effects across various cancers.
Collapse
Affiliation(s)
- Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India (A.P.)
| | - Deepanjali Kumar
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India (A.P.)
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India (A.P.)
| | - Madan Mohan Gupta
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
- Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur 303121, India
| |
Collapse
|
12
|
Ramakrishnan P, Ramprasath R, Jalaludeen AM, Jayakumar R, Jolius G, Balu R, Mohamed SB, Sridhar TM, Gunasekaran SS, Davoodbasha M, Thajuddin N, Gnanasekaran L, Sundaram T. Electrospun nanofibers of collagen and chitosan for tissue engineering and drug delivery applications: A review. Int J Biol Macromol 2025; 296:139663. [PMID: 39793786 DOI: 10.1016/j.ijbiomac.2025.139663] [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: 06/01/2024] [Revised: 12/16/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Tissue engineering plays a vital role in the medical field that addresses the repair, regeneration, and replacement of damaged tissues or organs. The development of drug-eluting electrospun nanofiber composed of biological macromolecules plays a key role in providing localized drug delivery and structural support. This review examines the recent development and impact of electrospun nanofibers in the field of tissue engineering and explores their potential applications. This review also investigates into the fabrication techniques of nanofibers, highlighting the use of biopolymers like collagen and chitosan, chiefly, focuses on understanding the mechanisms of drug-releasing features of these nanofibers. Studies concerning the medical applications of these nanofibers, such as wound healing, skin regeneration, bone tissue engineering, and neural repair, were also reviewed. Beyond the application in tissue regeneration, this review also explores the potential efficacy of nanofibres in cancer therapy, antibacterial activity, enzyme immobilization, and biosensing applications. This study provides an up-to-date critical insight into the applications of electrospun nanofiber application and key scalable production processes, underscoring the potential economic impacts of advanced wound care technologies. While outlining current challenges, this paper also offers future perspectives on the design, application, and potential expansion of drug-eluting electrospun fibers in medical sciences, ultimately showcasing their pivotal role in advancing therapeutic outcomes.
Collapse
Affiliation(s)
- Praveen Ramakrishnan
- Crescent Global Outreach Mission Research and Development, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai 620048, Tamil Nadu, India.
| | - Ramakrishnan Ramprasath
- Abinnovus Consulting Private Limited, TBI-University of Madras, Chennai 600025, Tamil Nadu, India
| | - Abdulkadhar Mohamed Jalaludeen
- Crescent Global Outreach Mission Research and Development, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai 620048, Tamil Nadu, India
| | - R Jayakumar
- Abinnovus Consulting Private Limited, TBI-University of Madras, Chennai 600025, Tamil Nadu, India
| | - Gimbun Jolius
- Center for Research in Advanced Fluid and Processes (Fluid Centre), Universiti Malaysia Pahang Al-Sultan Abdullah, 26300 Gambang, Pahang, Malaysia
| | - Ranganathan Balu
- CanBrs Therapeutics Private Limited, Indian Institute of Technology Madras - Research Park, Chennai, Tamil Nadu, India
| | - S B Mohamed
- Department of Materials Science, School of Technology, Central University of Tamil Nadu, Thiruvarur 610005, Tamil Nadu, India
| | - T M Sridhar
- Department of Analytical Chemistry, University of Madras, Chennai 600025, Tamil Nadu, India
| | - Sivagaami Sundari Gunasekaran
- Crescent Global Outreach Mission Research and Development, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai 620048, Tamil Nadu, India
| | - MubarakAli Davoodbasha
- Crescent Global Outreach Mission Research and Development, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai 620048, Tamil Nadu, India
| | - Nooruddin Thajuddin
- Crescent Global Outreach Mission Research and Development, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai 620048, Tamil Nadu, India
| | | | - Thanigaivel Sundaram
- Department of Biotechnology, Faculty of Science & Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu district, Tamil Nadu 603203, India.
| |
Collapse
|
13
|
Santos LS, Tonel MZ, Martins MO, dos Santos CL. Theoretical Exploration of Chitosan Nanoparticles Associated with Platinum Compounds for Cancer Treatment: Insights from DFT and Molecular Docking Analyses. BIONANOSCIENCE 2025; 15:79. [DOI: 10.1007/s12668-024-01728-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 01/04/2025]
|
14
|
Shahzad M, Hameed H, Amjad A, Khan MA, Qureshi IS, Hameed A, Saeed A, Munir R. An updated landscape on nanopharmaceutical delivery for mitigation of colon cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2107-2125. [PMID: 39361171 DOI: 10.1007/s00210-024-03482-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/21/2024] [Indexed: 03/19/2025]
Abstract
Globally, colorectal cancer (CRC) continues to rank among the leading causes of cancer-related death. Systemic toxicity, multidrug resistance, and nonspecific targeting often pose challenges to conventional therapy for CRC. Because it is a complex disease with a complex genetic and environmental pathophysiology, advanced therapeutic strategies are needed. Nanotechnology presents a potential solution that may maximize therapeutic efficacy while minimizing negative effects by enabling personalized delivery of anticancer drugs. This review focuses on recent developments in colorectal drug delivery systems based on nanotechnology. Numerous nanomaterials, including liposomes, dendrimers, micelles, exosomes, and gold nanoparticles, are developed and used. Distinctive characteristics of mentioned nanocarriers are discussed along with strategies that can be employed for enhancing the delivery of drugs to colorectal cancer cells. The review also quotes the most relevant preclinical and clinical studies that show how these nanomaterials improve drug solubility, stability, and targeted delivery while overcoming the shortcomings of conventional therapies. Nanotechnology has made CRC treatment very efficient and advanced, which has opened up new possibilities for targeted drug delivery. Preclinical and clinical studies have also proved that the use of nano-formulations in colon-specific delivery systems have significant results, indicating potential for better patient outcomes. Future research can be done in order to overcome the hurdles regarding biocompatibility, expansion, and regulatory challenges. Large-scale clinical trials and nanomaterial formulation optimization should be the main goals of future research to confirm the efficacy and safety of these novel treatments.
Collapse
Affiliation(s)
- Maria Shahzad
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Ayesha Amjad
- Faculty of Food Technology and Nutrition Sciences, Lahore University of Biological and Applied Sciences, Lahore, 54000, Pakistan
| | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Inaba Shujaat Qureshi
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan
| | - Asad Saeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan
| | - Rabia Munir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| |
Collapse
|
15
|
Esmaeili Z, Shavali Gilani P, Khosravani M, Motamedi M, Maleknejad S, Adabi M, Sadighara P. Nanotechnology-driven EGCG: bridging antioxidant and therapeutic roles in metabolic and cancer pathways. Nanomedicine (Lond) 2025; 20:621-636. [PMID: 39924937 PMCID: PMC11881875 DOI: 10.1080/17435889.2025.2462521] [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: 10/28/2024] [Accepted: 01/31/2025] [Indexed: 02/11/2025] Open
Abstract
Epigallocatechin-3-gallate (EGCG), the primary polyphenol in green tea, is renowned for its potent antioxidant properties. EGCG interacts with various cellular targets, inhibiting cancer cell proliferation through apoptosis and cell cycle arrest induction, while also modulating metabolic pathways. Studies have demonstrated its potential in addressing cancer development, obesity, and diabetes. Given the rising prevalence of metabolic diseases and cancers, EGCG is increasingly recognized as a promising therapeutic agent. This review provides a comprehensive overview of the latest findings on the effects of both free and nano-encapsulated EGCG on mechanisms involved in the management and prevention of hyperlipidemia, diabetes, and gastrointestinal (GI) cancers. The review highlights EGCG role in modulating key signaling pathways, enhancing bioavailability through nano-formulations, and its potential applications in clinical settings.
Collapse
Affiliation(s)
- Zahra Esmaeili
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Shavali Gilani
- Department of Environmental Health Engineering, Division of Food Safety and Hygiene, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maral Motamedi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shokofeh Maleknejad
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Sadighara
- Department of Environmental Health Engineering, Division of Food Safety and Hygiene, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Nyandoro VO, Ismail EA, Tageldin A, Gafar MA, Peters XQ, Mautsoe R, Omolo CA, Govender T. Potential of nanocarrier-mediated delivery of vancomycin for MRSA infections. Expert Opin Drug Deliv 2025; 22:347-365. [PMID: 39949087 DOI: 10.1080/17425247.2025.2459756] [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: 10/28/2024] [Accepted: 01/24/2025] [Indexed: 02/20/2025]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) threatens global health due to its resistance to vancomycin, which is the standard treatment despite limitations, including nephrotoxicity and low intracellular permeability. This necessitates the development of innovative strategies such as nanocarrier-mediated delivery to overcome such limitations. Nanocarriers serve as delivery systems for vancomycin and exhibit inherent antibacterial properties, potentially providing synergism and overcoming MRSA's resistance. Nanocarriers provide sustained release and targeted delivery of vancomycin to the infection site, achieving higher therapeutic concentrations and superior antibacterial activity with reduced doses, which minimizes systemic toxicity. Moreover, leveraging simulations techniques provides more insights on vancomycin-nanocarrier interactions, facilitating the optimization of nanosystems. AREAS COVERED The article discusses the potential of nanocarriers in delivering vancomycin to infection site, reducing systemic toxicity, and potentiating anti-MRSA activity. Additionally, it reviews modeling and simulation studies to provide a deeper understanding of vancomycin-nanocarrier interactions. The literature search included experimental articles from 2017 to 2024, searched in Web of Science, Google scholar, PubMed, and Scopus. EXPERT OPINION Nanocarrier-mediated delivery of vancomycin offers promising approaches to combat MRSA infections by enhancing therapeutic efficacy and reducing systemic toxicity. However, further research is required to optimize these nanoformulations and advance them to clinical trials and practical applications.
Collapse
Affiliation(s)
- Vincent O Nyandoro
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Pharmaceutics and Pharmaceutical Chemistry, School of Pharmacy, Kabarak University, Kabarak, Kenya
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Medani, Sudan
| | - Abdelrahman Tageldin
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Mohammed A Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Xylia Q Peters
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Relebohile Mautsoe
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- School of Pharmacy and Health Sciences, Department of Pharmaceutics, United States International University-Africa, Nairobi, Kenya
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
17
|
Moumivand A, Naderi F, Moradi O, Makiabadi B. Smart drug delivery: a DFT study of C 24 fullerene and doped analogs for pyrazinamide. NANOSCALE ADVANCES 2025; 7:1287-1299. [PMID: 39802333 PMCID: PMC11712202 DOI: 10.1039/d4na00560k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
The potential applicability of the C24 nanocage and its boron nitride-doped analogs (C18B3N3 and C12B6N6) as pyrazinamide (PA) carriers was investigated using density functional theory. Geometry optimization and energy calculations were performed using the B3LYP functional and 6-31G(d) basis set. Besides, dispersion-corrected interaction energies were calculated at CAM (Coulomb attenuated method)-B3LYP/6-31G(d,p) and M06-2X/6-31G(d,p) levels of theory. The adsorption energy (E ads), enthalpy (ΔH), and Gibbs free energy (ΔG) values for C24-PA, C18B3N3-PA, and C12B6N6-PA structures were calculated. The molecular descriptors such as electrophilicity (ω), chemical potential (μ), chemical hardness (η) and chemical softness (S) of compounds were investigated. Natural bond orbital (NBO) analysis confirms the charge transfer from the drug molecule to nanocarriers upon adsorption. Based on the quantum theory of atoms in molecules (QTAIM), the nature of interactions in the complexes was determined. These findings suggest that C24 and its doped analogs are promising candidates for smart drug delivery systems and PA sensing applications, offering significant potential for advancements in targeted tuberculosis treatment.
Collapse
Affiliation(s)
- Azam Moumivand
- Department of Chemistry, Shahr-e-Qods Branch, Islamic Azad University Tehran Iran https://scholar.google.com/citations?user=pdCrLOYAAAAJ&hl=en
| | - Fereshteh Naderi
- Department of Chemistry, Shahr-e-Qods Branch, Islamic Azad University Tehran Iran https://scholar.google.com/citations?user=pdCrLOYAAAAJ&hl=en
| | - Omid Moradi
- Department of Chemistry, Shahr-e-Qods Branch, Islamic Azad University Tehran Iran https://scholar.google.com/citations?user=pdCrLOYAAAAJ&hl=en
| | - Batoul Makiabadi
- Department of Chemical Engineering, Sirjan University of Technology Sirjan Iran https://scholar.google.com/citations?user=N6z-rHsAAAAJ&hl=en
| |
Collapse
|
18
|
Martins M, Veiga F, Paiva-Santos AC, Pires PC. Drug Repurposing and Nanotechnology for Topical Skin Cancer Treatment: Redirecting toward Targeted and Synergistic Antitumor Effects. ACS Pharmacol Transl Sci 2025; 8:308-338. [PMID: 39974652 PMCID: PMC11833728 DOI: 10.1021/acsptsci.4c00679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Skin cancer represents a major health concern due to its rising incidence and limited treatment options. Current treatments (surgery, chemotherapy, radiotherapy, immunotherapy, and targeted therapy) often entail high costs, patient inconvenience, significant adverse effects, and limited therapeutic efficacy. The search for novel treatment options is also marked by the high capital investment and extensive development involved in the drug discovery process. In response to these challenges, repurposing existing drugs for topical application and optimizing their delivery through nanotechnology could be the answer. This innovative strategy aims to combine the advantages of the known pharmacological background of commonly used drugs to expedite therapeutic development, with nanosystem-based formulations, which among other advantages allow for improved skin permeation and retention and overall higher therapeutic efficacy and safety. The present review provides a critical analysis of repurposed drugs such as doxycycline, itraconazole, niclosamide, simvastatin, leflunomide, metformin, and celecoxib, formulated into different nanosystems, namely, nanoemulsions and nanoemulgels, nanodispersions, solid lipid nanoparticles, nanostructured lipid carriers, polymeric nanoparticles, hybrid lipid-polymer nanoparticles, hybrid electrospun nanofibrous scaffolds, liposomes and liposomal gels, ethosomes and ethosomal gels, and aspasomes, for improved outcomes in the battle against skin cancer. Enhanced antitumor effects on melanoma and nonmelanoma research models are highlighted, with some nanoparticles even showing intrinsic anticancer properties, leading to synergistic effects. The explored research findings highly evidence the potential of these approaches to complement the currently available therapeutic strategies in the hope that these treatments might one day reach the pharmaceutical market.
Collapse
Affiliation(s)
- Maria Martins
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Patrícia C. Pires
- Department
of Pharmaceutical Technology, Faculty of
Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV,
Group of Pharmaceutical Technology, Faculty
of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- RISE-Health,
Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
19
|
Pyrczak-Felczykowska A, Herman-Antosiewicz A. Modification in Structures of Active Compounds in Anticancer Mitochondria-Targeted Therapy. Int J Mol Sci 2025; 26:1376. [PMID: 39941144 PMCID: PMC11818413 DOI: 10.3390/ijms26031376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer is a multifaceted disease characterised by uncontrolled cellular proliferation and metastasis, resulting in significant global mortality. Current therapeutic strategies, including surgery, chemotherapy, and radiation therapy, face challenges such as systemic toxicity and tumour resistance. Recent advancements have shifted towards targeted therapies that act selectively on molecular structures within cancer cells, reducing off-target effects. Mitochondria have emerged as pivotal targets in this approach, given their roles in metabolic reprogramming, retrograde signalling, and oxidative stress, all of which drive the malignant phenotype. Targeting mitochondria offers a promising strategy to address these mechanisms at their origin. Synthetic derivatives of natural compounds hold particular promise in mitochondrial-targeted therapies. Innovations in drug design, including the use of conjugates and nanotechnology, focus on optimizing these compounds for mitochondrial specificity. Such advancements enhance therapeutic efficacy while minimizing systemic toxicity, presenting a significant step forward in modern anticancer strategies.
Collapse
Affiliation(s)
| | - Anna Herman-Antosiewicz
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, 80-308 Gdańsk, Poland;
| |
Collapse
|
20
|
Aarthi J, Kaviya S, Kirubavathi K, Selvaraju K, Shakeel F, Gowri S, Faiyazuddin M. Green synthesis of nickel oxide nanoparticles using leaf extract of Mimosa pudica for killing triple-negative breast cancer MDA-MB-231 cells and bacteria. J Mol Struct 2025; 1321:140178. [DOI: 10.1016/j.molstruc.2024.140178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
21
|
Zhang Z, Wu H, Li M, Zhou F, Huang Y. From natural herbal wisdom to nano innovation: Revolutionizing tumor treatment through intervening in metabolic reprogramming. Biochim Biophys Acta Rev Cancer 2025; 1880:189263. [PMID: 39800231 DOI: 10.1016/j.bbcan.2025.189263] [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: 11/03/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
In recent years, with the deepening understanding of the biological mechanisms underlying tumorigenesis, metabolic reprogramming has emerged as a pivotal process in cancer initiation, progression, and treatment resistance, gradually paving the way for new avenues in precision oncology. Natural herbal ingredients, characterized by their multi-target engagement, low toxicity, and wide-ranging biological activities, exhibit unique advantages in anti-cancer therapy. Nonetheless, the clinical application of these components has been constrained by issues such as poor solubility, low bioavailability, and inadequate stability when administered through traditional delivery methods. The advent of multifunctional nanoformulations has offered solutions to these challenges, substantially advancing the utilization of natural herbal components in precision therapy targeting tumor metabolic reprogramming. This article provides a comprehensive review of the multidimensional features of cancer metabolic reprogramming and its intricate regulatory network, highlighting the latest advancements in metabolic regulation, targeted delivery, and precision therapy achieved through natural herbs and their multifunctional nanomedicines. It also offers insights into future directions in this field. We are justified in believing that continued breakthroughs in this area will usher in safer and more effective treatment options for cancer patients, heralding a new chapter in cancer therapy.
Collapse
Affiliation(s)
- Zhengguang Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China; School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Haitao Wu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| | - Yan Huang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, China.
| |
Collapse
|
22
|
Repetowski P, Warszyńska M, Dąbrowski JM. NIR-activated multifunctional agents for the combined application in cancer imaging and therapy. Adv Colloid Interface Sci 2025; 336:103356. [PMID: 39612723 DOI: 10.1016/j.cis.2024.103356] [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: 08/16/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Anticancer therapies that combine both diagnostic and therapeutic capabilities hold significant promise for enhancing treatment efficacy and patient outcomes. Among these, agents responsive to near-infrared (NIR) photons are of particular interest due to their negligible toxicity and multifunctionality. These compounds are not only effective in photodynamic therapy (PDT), but also serve as contrast agents in various imaging modalities, including fluorescence and photoacoustic imaging. In this review, we explore the photophysical and photochemical properties of NIR-activated porphyrin, cyanine, and phthalocyanines derivatives as well as aggregation-induced emission compounds, highlighting their application in synergistic detection, diagnosis, and therapy. Special attention is given to the design and optimization of these agents to achieve high photostability, efficient NIR absorption, and significant yields of fluorescence, heat, or reactive oxygen species (ROS) generation depending on the application. Additionally, we discuss the incorporation of these compounds into nanocarriers to enhance their solubility, stability, and target specificity. Such nanoparticle-based systems exhibit improved pharmacokinetics and pharmacodynamics, facilitating more effective tumor targeting and broadening the application range to photoacoustic imaging and photothermal therapy. Furthermore, we summarize the application of these NIR-responsive agents in multimodal imaging techniques, which combine the advantages of fluorescence and photoacoustic imaging to provide comprehensive diagnostic information. Finally, we address the current challenges and limitations of photodiagnosis and phototherapy and highlight some critical barriers to their clinical implementation. These include issues related to their phototoxicity, limited tissue penetration, and potential off-target effects. The review concludes by highlighting future research directions aimed at overcoming these obstacles, with a focus on the development of next-generation agents and platforms that offer enhanced therapeutic efficacy and imaging capabilities in the field of cancer treatment.
Collapse
Affiliation(s)
- Paweł Repetowski
- Faculty of Chemistry, Jagiellonian University, 30-387 Kraków, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Kraków, Poland
| | - Marta Warszyńska
- Faculty of Chemistry, Jagiellonian University, 30-387 Kraków, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Kraków, Poland
| | | |
Collapse
|
23
|
Li T, Cheng D, Xu X, Wang B, Xing W, Xu Y, Qian X, Yang Y, Zhu W. Transferrin-targeting pH-responsive and biodegradable mesoporous silica nanohybrid for nitric oxide-sensitized chemotherapy of cancer. Colloids Surf B Biointerfaces 2025; 246:114409. [PMID: 39612521 DOI: 10.1016/j.colsurfb.2024.114409] [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: 08/21/2024] [Revised: 11/16/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
Weakly acidic pH, low oxygen and high glutathione levels are the main characteristics of tumor cells. Taking advantage of the unique acidic microenvironment of tumor cells, acid-responsive mesoporous organosilica nanoparticles (AMON) were designed for nitric oxide (NO)-sensitized chemotherapy of tumors. AMON served as a nanocarrier co-loaded with a nitric oxide donor (NOD) and chemotherapeutic drug doxorubicin (DOX). Transferrin (Tf) was modified on the surface as a targeting ligand to form NOD&DOX@AMON. In vitro experiments showed that AMON could be completely degraded under acidic conditions (pH 5.0) after 48 h. NOD&DOX@AMON entered cells via transferrin receptor-mediated internalization and degraded in the acidic microenvironment to release its payloads. NOD released NO in presence of one-electron reducing substances like Glutathione (GSH) and ascorbic acid, inhibiting P-glycoprotein(P-gp) function and thereby increasing the intracellular concentration of DOX. In vivo distribution studies revealed that the nanohybrids accumulated maximally in tumor tissue 12 h after intravenous injection and exhibited significant inhibitory effects on HepG2 xenograft tumors. Western blot experiments demonstrated that NOD&DOX@AMON could inhibit the expression of drug resistance-associated proteins and was expected to be employed as a therapeutic approach for drug-resistant ttumors.
Collapse
Affiliation(s)
- Ting Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Di Cheng
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiu Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Bin Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wenqian Xing
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yufang Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xuhong Qian
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Youjun Yang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weiping Zhu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
24
|
Famta P, Shah S, Vambhurkar G, Pandey G, Bagasariya D, Kumar KC, Prasad SB, Shinde A, Wagh S, Srinivasarao DA, Kumar R, Khatri DK, Asthana A, Srivastava S. Amelioration of breast cancer therapies through normalization of tumor vessels and microenvironment: paradigm shift to improve drug perfusion and nanocarrier permeation. Drug Deliv Transl Res 2025; 15:389-406. [PMID: 39009931 DOI: 10.1007/s13346-024-01669-9] [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] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer among women. Chemo-, immune- and photothermal therapies are employed to manage BC. However, the tumor microenvironment (TME) prevents free drugs and nanocarriers (NCs) from entering the tumor premises. Formulation scientists rely on enhanced permeation and retention (EPR) to extravasate NCs in the TME. However, recent research has demonstrated the inconsistent nature of EPR among different patients and tumor types. In addition, angiogenesis, high intra-tumor fluid pressure, desmoplasia, and high cell and extracellular matrix density resist the accumulation of NCs in the TME. In this review, we discuss TME normalization as an approach to improve the penetration of drugs and NCSs in the tumor premises. Strategies such as normalization of tumor vessels, reversal of hypoxia, alleviation of high intra-tumor pressure, and infiltration of lymphocytes for the reversal of therapy failure have been discussed in this manuscript. Strategies to promote the infiltration of anticancer immune cells in the TME after vascular normalization have been discussed. Studies strategizing time points to administer TME-normalizing agents are highlighted. Mechanistic pathways controlling the angiogenesis and normalization processes are discussed along with the studies. This review will provide greater tumor-targeting insights to the formulation scientists.
Collapse
Affiliation(s)
- Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Deepkumar Bagasariya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Kondasingh Charan Kumar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Sajja Bhanu Prasad
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Akshay Shinde
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Suraj Wagh
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
- Molecular and Cellular Biology Laboratory, Department of Pharmacology, Nims Institute of Pharmacy, Nims University, Jaipur, Rajasthan, India
| | - Amit Asthana
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
25
|
He M, Chen S, Yu H, Fan X, Wu H, Wang Y, Wang H, Yin X. Advances in nanoparticle-based radiotherapy for cancer treatment. iScience 2025; 28:111602. [PMID: 39834854 PMCID: PMC11743923 DOI: 10.1016/j.isci.2024.111602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Radiotherapy has long been recognized as an effective conventional approach in both clinical and scientific research, primarily through mechanisms involving DNA destruction or the generation of reactive oxygen species to target tumors. However, significant challenges persist, including the unavoidable damage to normal tissues and the development of radiation resistance. As a result, nanotechnology-based radiotherapy has garnered considerable attention for its potential to enhance precision in irradiation, improve radiosensitization, and achieve therapeutic advancements. Importantly, radiotherapy alone frequently falls short of fully eradicating tumors. Consequently, to augment the efficacy of radiotherapy, it is often integrated with other therapeutic strategies. This review elucidates the mechanisms of radiotherapy sensitization based on diverse nanoparticles. Typically, radiotherapy is sensitized through augmenting reactive oxygen species production, targeted radiotherapy, hypoxia relief, enhancement of antitumor immune microenvironment, and G2/M cell cycle arrest. Moreover, the incorporation of nanoparticle-based anti-tumor strategies with radiotherapy markedly enhances the current state of radiotherapy. Additionally, a compilation of clinical trials utilizing nano-radioenhancers is presented. Finally, future prospects for clinical translation in this field are thoroughly examined.
Collapse
Affiliation(s)
- Meijuan He
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shixiong Chen
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
| | - Hongwei Yu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xuhui Fan
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hong Wu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yihui Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
| | - Han Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
- Jiading Branch of Shanghai General Hospital, Shanghai 201803, China
| | - Xiaorui Yin
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
26
|
Zhang Z, Wang R, Chen L. Drug Delivery System Targeting Cancer-Associated Fibroblast for Improving Immunotherapy. Int J Nanomedicine 2025; 20:483-503. [PMID: 39816375 PMCID: PMC11734509 DOI: 10.2147/ijn.s500591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous population of non-malignant cells that play a crucial role in the tumor microenvironment, increasingly recognized as key contributors to cancer progression, metastasis, and treatment resistance. So, targeting CAFs has always been considered an important part of cancer immunotherapy. However, targeting CAFs to improve the efficacy of tumor therapy is currently a major challenge. Nanomaterials show their unique advantages in the whole process. At present, nanomaterials have achieved significant accomplishments in medical applications, particularly in the field of cancer-targeted therapy, showing enormous potential. It has been confirmed that nanomaterials can not only directly target CAFs, but also interact with the tumor microenvironment (TME) and immune cells to affect tumorigenesis. As for the cancer treatment, nanomaterials could enhance the therapeutic effect in many ways. Therefore, in this review, we first summarized the current understanding of the complex interactions between CAFs and TME, immune cells, and tumor cells. Next, we discussed common nanomaterials in modern medicine and their respective impacts on the TME, CAFs, and interactions with tumors. Finally, we focus on the application of nano drug delivery system targeting CAFs in cancer therapy.
Collapse
Affiliation(s)
- Zhongsong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| | - Rong Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| | - Long Chen
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| |
Collapse
|
27
|
Saghiri MA, Saini RS, Heboyan A. Cytotoxicity of Nanocarrier-Based Drug Delivery in Oral Cancer Therapy: A Systematic Review and Meta-Analysis. Cancer Control 2025; 32:10732748241310936. [PMID: 39829067 PMCID: PMC11744631 DOI: 10.1177/10732748241310936] [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: 10/28/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Oral cancer remains 1 of the biggest health care challenges; it has a poor response to treatment, and treatment often results in severe side effects. Nano-targeted drug carrier-assisted drug delivery systems can improve the benefits of targeted drug delivery and treatment efficacy. A systematic review and meta-analysis was conducted to investigate the effect of targeted nano carrier drug delivery systems on the management of oral cancer. METHODS A comprehensive literature search was performed using PubMed, ScienceDirect, the Cochrane Library, Google Scholar, and Scopus using PRISMA guidelines, to identify relevant in vitro and in vivo (human) studies. Studies evaluating the impact of nanocarrier-based delivery systems on oral cancer cells or human models were selected. Pooled effect sizes were calculated using random-effects models via RevMan 5.4, and heterogeneity among studies was assessed. RESULTS After full-text assessment, 15 research articles were included [14 in vitro studies and 1 randomized controlled trial (RCT)]. In the meta-analysis, the pooled data (IC50) for the impact of the nanocarrier delivery system vs control on oral cancer was -7.67 (95% CI: -41.77, 26.43), with a high heterogeneity (I2 = 92%, P < 0.00001). Moreover, in vitro studies had a medium risk of bias, while the RCT had some concerns in the randomization domain. CONCLUSION Nanocarrier-based drug delivery has been found to be a superior approach compared to drug delivery in free form, increasing the efficacy and safety of oral cancer treatment.
Collapse
Affiliation(s)
- Mohammad A. Saghiri
- Department of Restorative Dentistry, Director of Biomaterial and Prosthodontic Laboratory, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - Ravinder S. Saini
- Department A. Dental Health Sciences COAMS, King Khalid University, Abha, Saudi Arabia
| | - Artak Heboyan
- Department of Prosthodontics, Faculty of Stomatology, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia
| |
Collapse
|
28
|
Prajapati M, Deshmukh R, Harwansh RK. Recent Trends in Nanoparticulate Delivery System for Amygdalin as Potential Therapeutic Herbal Bioactive Agent for Cancer Treatment. Curr Drug Deliv 2025; 22:63-79. [PMID: 38037911 DOI: 10.2174/0115672018280381231119150732] [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: 09/02/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023]
Abstract
Cancer is the deadliest and most serious health problem. The mortality rate of cancer patients has increased significantly worldwide in recent years. There are several treatments available, but these treatments have many limitations, such as non-specific targeting, toxicity, bioavailability, solubility, permeability problems, serious side effects, and a higher dose. Many people prefer phytomedicine because it has fewer side effects. However, amygdalin is a naturally occurring phytoconstituent. It has many harmful effects due to the cyanide group present in the chemical structure. Many scientists and researchers have given their thoughts associated with amygdalin and its toxicities. However, there is a need for a more advanced, effective, and newer delivery system with reduced toxicity effects of amygdalin. Nanotechnology has become a more refined and emerging medical approach, offering innovative research areas to treat cancer. This review focuses on the use of amygdaline as herbal medicine encapsulating into several nanoparticulate delivery systems such as silver nanoparticles, graphene oxide nanoparticles, gold nanoparticles, nanofibers, nanocomposites, niosomes, and magnetic nanoparticles in the treatment of cancer. In addition, this article provides information on amygdalin structure and physical properties, pharmacokinetics, toxicity, and challenges with amygdalin.
Collapse
Affiliation(s)
- Mahendra Prajapati
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| |
Collapse
|
29
|
Dwivedi K, Sahoo A, Almalki WH, Almujri SS, Aodah A, Alruwaili NK, Rab SO, Alanezi AA, Haji EM, Barkat MA, Singh T, Rahman M. Innovative nanocarrier systems for enhanced delivery of phyto-active compounds in cancer therapy. Nanomedicine (Lond) 2025; 20:91-116. [PMID: 39703154 DOI: 10.1080/17435889.2024.2440301] [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: 07/27/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
Millions of people worldwide suffer from cancer, facing challenges such as treatments affecting healthy cells, suboptimal responses, adverse effects, recurrence risk, drug resistance, and nonspecific targeting. Chemoresistance leads to fatalities, but phytoactives show promise in cancer management despite limitations such as high metabolism, poor absorption, and high dosage requirements. Challenges in the large-scale isolation of phytoactive compounds, solubility, bioavailability, and targeting limit their development. Recent developments, including carbohydrate, lipid, and protein-based nanoparticles, have enhanced cancer treatment by improving the bioavailability and targeted delivery of phytoactives such as polyphenols, alkaloids, sulfur-containing compounds, flavonoids, and terpenes. Despite advancements, clinical application faces hurdles such as poor bioavailability and inconsistent immune responses. This article discusses the promise of phytoactive-loaded nanoformulations in cancer management, highlighting targeted drug delivery, unmet needs, and challenges. Further research is needed to overcome these challenges and fully understand the potential of phytoactives in cancer management.
Collapse
Affiliation(s)
- Khusbu Dwivedi
- Department of Pharmaceutics, Shambhunath Institute of Pharmacy, Prayagraj, India
| | - Ankit Sahoo
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Asir-Abha, Saudi Arabia
| | - Alhussain Aodah
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Nabil K Alruwaili
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Science, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Abdulkareem Ali Alanezi
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al Batin, Saudi Arabia
| | - Esraa M Haji
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al Batin, Saudi Arabia
| | - Tanuja Singh
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Mahfoozur Rahman
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| |
Collapse
|
30
|
Priya, Gaur PK, Kumar S. Nanocarrier-Mediated Dermal Drug Delivery System of Antimicrobial Agents for Targeting Skin and Soft Tissue Infections. Assay Drug Dev Technol 2025; 23:2-28. [PMID: 39587945 DOI: 10.1089/adt.2024.060] [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: 11/27/2024] Open
Abstract
Antimicrobial resistance in disease-causing microbes is seen as a severe problem that affects the entire world, makes therapy less effective, and raises mortality rates. Dermal antimicrobial therapy becomes a desirable choice in the management of infectious disorders since the rising resistance to systemic antimicrobial treatment frequently necessitates the use of more toxic drugs. Nanoparticulate systems such as nanobactericides, which have built-in antibacterial activity, and nanocarriers, which function as drug delivery systems for conventional antimicrobials, are just two examples of the treatment methods made feasible by nanotechnology. Silver nanoparticles, zinc oxide nanoparticles, and titanium dioxide nanoparticles are examples of inorganic nanoparticles that are efficient on sensitive and multidrug-resistant bacterial strains both as nanobactericides and nanocarriers. To stop the growth of microorganisms that are resistant to standard antimicrobials, various antimicrobials for dermal application are widely used. This review covers the most prevalent microbes responsible for skin and soft tissue infections, techniques to deliver dermal antimicrobials, topical antimicrobial safety concerns, current issues, challenges, and potential future developments. A thorough and methodical search of databases, such as Google Scholar, PubMed, Science Direct, and others, using specified keyword combinations, such as "antimicrobials," "dermal," "nanocarriers," and numerous others, was used to gather relevant literature for this work.
Collapse
Affiliation(s)
- Priya
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology, Meerut, Uttar Pradesh, India
| | - Praveen Kumar Gaur
- Department of Pharmaceutics, Metro College of Health Sciences & Research, Greater Noida, Uttar Pradesh, India
| | - Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology, Meerut, Uttar Pradesh, India
| |
Collapse
|
31
|
Zarrinnahad H, Dehdast SA, Fard GC, Nourbakhsh M, Koohi MK, Panahi G, Karimpour A, Rezayat SM, Shabani M. The effect of biosynthesized zinc oxide nanoparticles on gene expression and apoptosis in triple-negative breast cancer cells. Daru 2024; 33:10. [PMID: 39731629 PMCID: PMC11682029 DOI: 10.1007/s40199-024-00553-8] [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: 07/10/2024] [Accepted: 10/25/2024] [Indexed: 12/30/2024] Open
Abstract
OBJECTIVE(S) Some forms of breast cancer such as triple-negative phenotype, are serious challenge because of high metastatic cases, high mortality and resistance to conventional therapy motivated the search for alternative treatment approaches. Nanomaterials are promising candidates and suitable alternatives for improving tumor and cancer cell treatments. MATERIALS AND METHODS Biosynthesis of ZnO NPs by help of Berberis integerrima fruit extract, has been done. Analysis of Zinc Oxide NPs using DLS, FTIR, SEM, and EDS techniques have been performed. Moreover, biological activities of ZnO NPs evaluated through MTT method, Flow cytometry, and real time PCR methods. Biocatalytic and apoptotic activity of ZnO NPs on healthy HFF (human fibroblast cell line), MDA-MB 231, and MDA-MB 468 (triple negative breast cancer cell lines, (TNBC)) evaluated. Furthermore, Bax, Bcl-2 and caspase-3 apoptotic genes expression changes in cancer cells assessed in compare to GAPDH as a house keeping gene. RESULTS Physico-chemical investigation demonstrated ZnO NPs were confirmed by Berberis integerrima fruit extract for the first time. The MTT assay and Flow cytometry results indicated biocompatibility of the ZnO NPs in normal cell line and high anticancer potential against TNBC MDA-MB-231 and MDA-MB-468 cell lines. The IC50 of ZnO NPs were 104.4 and 44.86, 20.96 after 24 hours for HFF, MDA-MB-231 and MDA-MB-468 cells, respectively. CONCLUSION The current research showed a fast, cost effective and ecofriendly method for ZnO NPs nanoparticle synthesis. Furthermore, In vitro data analysis demonstrated biocompatibility and highly anticancer effects of biosynthesized ZnO NPs against TNBC cancerous cells.
Collapse
Affiliation(s)
- Hannaneh Zarrinnahad
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - S Ahmad Dehdast
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Chizari Fard
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Clothing and Fabric Design Department, Art Faculty, Imam Javad University College, Yazd, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Kazem Koohi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ghodratollah Panahi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Karimpour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - S Mehdi Rezayat
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Shabani
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Kumar G, Virmani T, Chhabra V, Virmani R, Pathak K, Akhtar MS, Hussain Asim M, Arshad S, Siddique F, Fonte P. Transforming cancer treatment: The potential of nanonutraceuticals. Int J Pharm 2024; 667:124919. [PMID: 39515676 DOI: 10.1016/j.ijpharm.2024.124919] [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: 06/06/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Chemotherapy in the management of cancer is constrained by limitations like off-target effects, poor bioavailability, and dose-dependent toxicity. Nutraceuticals have been explored as an innovative strategy to overcome chemotherapy drawbacks.However, the clinical utility of nutraceuticals is restricted due to their complex structures, less water solubility, reduced stability, decreased bioavailability and more obstacles in the gastrointestinal tract. Nanonutraceuticals are nanosized nutraceutical particles having enhanced solubility, improved bioavailability, stability, and targeted delivery to specific cells. Nutraceuticals can be co-delivered with other chemotherapeutic drugs in nanocarriers to elicit synergistic effects. The targeting of nutraceuticals against cancer cells can be enabled by coupling ligands with the nanocarriers, which direct to the overexpressed receptors found at the surface of the cancer cells. Transitioning a nanonutraceutical from pre-clinical research to clinical trials is a pivotal step. This focus on advancing their application holds great potential for impacting clinical research and improving the treatment landscape for cancer patients. This review focuses on the role of nutraceuticals for cancer treatment, various nanocarriers for the efficient delivery of nutraceuticals along with co-administration of nutraceuticals with chemotherapeutic drugs using nanocarriers. Also, emphasize the targeting of ligands coupled nanocarriers to the cancer cells along with patents and clinical trials for nanonutraceuticals.
Collapse
Affiliation(s)
- Girish Kumar
- Amity Institute of Pharmacy, Amity University Greater Noida, Uttar Pradesh 201308, India
| | - Tarun Virmani
- Amity Institute of Pharmacy, Amity University Greater Noida, Uttar Pradesh 201308, India.
| | - Vaishnavi Chhabra
- National Institute of Pharmaceutical Education & Research, Mohali, Punjab 160062, India
| | - Reshu Virmani
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah, Uttar Pradesh 206001, India
| | - Md Sayeed Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, AlFara, Abha 62223, Saudi Arabia
| | | | - Shumaila Arshad
- Doctor's Institute of Health Sciences, 3-Km Sargodha Bypass Road, Sargodha 40100, Pakistan
| | - Farzana Siddique
- Institute of Food Science and Nutrition, University of Sargodha, Sargodha 40100, Pakistan
| | - Pedro Fonte
- Center for Marine Sciences (CCMAR), University of Algarve, Gambelas Campus, Faro 8005-139, Portugal; Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, University of Algarve, Gambelas Campus, Faro 8005-139, Portugal; iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal.
| |
Collapse
|
33
|
Rupel K, Fanfoni L, Dus J, Tommasini M, Porrelli D, Medagli B, Canfora F, Adamo D, Di Lenarda R, Ottaviani G, Biasotto M. Development and Characterization of Biocompatible Cellulose-Tetraphenylethylene Hydrazone Self-Assembling Nanomicelles with Acidity-Triggered Release of Doxorubicin for Cancer Therapy. Curr Issues Mol Biol 2024; 46:14244-14258. [PMID: 39727981 DOI: 10.3390/cimb46120853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
The development of anticancer diagnostic and therapeutic strategies is of crucial importance to improve efficacy and therapeutic specificity. Here, we describe the synthesis and characterization of fluorescent self-assembling nanomicelles (NMs) based on a biocompatible polysaccharide (cellulose, CE) functionalized with a tetraphenyl ethylene derivative (TPEHy) and loaded with Doxorubicin (DOX) with aggregation-induced emission (AIE) properties and pH-dependent drug release. We obtained CE-TPEHy-NMs with an average diameter of 60 ± 17 nm for unloaded NMs and 86 ± 25 nm for NMs loaded with DOX, respectively. Upon testing different conditions, we obtained an encapsulation efficiency of 86% and a loading capacity of 90%. A controlled dialysis experiment showed that the release of DOX after 48 h is minimal at pH 7.4 (11%), increasing at pH 6.5 (50%) and at its maximum at pH 4.5 (80%). The cytotoxicity of blank and loaded CE-TPEHy-NMs at increasing concentrations and different pH conditions was tested on a MG-63 human osteosarcoma cell line. Based on viability assays at pH 7.4, neither unloaded nor loaded CE-TPEHy-NMs exerted any inhibition on cell proliferation. At pH 6.5, proliferation inhibition significantly increased, confirming the pH-dependent release. We characterized and studied the performance of CE-based amphiphilic, biocompatible NMs for controlled drug release in acidic conditions, such as tumor microenvironments. Further studies are required to optimize their synthesis process and to validate their antitumoral properties in vivo.
Collapse
Affiliation(s)
- Katia Rupel
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Lidia Fanfoni
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Jacopo Dus
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Martina Tommasini
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Davide Porrelli
- Department of Life Sciences, University of Trieste, Via Alexander Fleming 31, 34127 Trieste, Italy
| | - Barbara Medagli
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Federica Canfora
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 5 Via Pansini, 80131 Naples, Italy
| | - Daniela Adamo
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 5 Via Pansini, 80131 Naples, Italy
| | - Roberto Di Lenarda
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Giulia Ottaviani
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Matteo Biasotto
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| |
Collapse
|
34
|
Younis MA, Alsogaihi MA, Abdellatif AAH, Saleem I. Nanoformulations in the treatment of lung cancer: current status and clinical potential. Drug Dev Ind Pharm 2024:1-17. [PMID: 39629952 DOI: 10.1080/03639045.2024.2437562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/17/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
OBJECTIVE Recent developments in nanotechnology have regained hope in enabling the eradication of lung cancer, while overcoming the drawbacks of the classic therapeutics. Nevertheless, there are still formidable obstacles that hinder the translation of such platforms from the bench into the clinic. Herein, we shed light on the clinical potential of these formulations and discuss their future directions. SIGNIFICANCE OF REVIEW The current article sheds light on the recent advancements in the recruitment of nanoformulations against lung cancer, focusing on their unique features, merits, and demerits. Moreover, inorganic nanoparticles, including gold, silver, magnetic, and carbon nanotubes are highlighted as emerging drug delivery technologies. Furthermore, the clinical status of these formulations is discussed, with particular attention on the challenges that they encounter in their clinical translation. Lastly, the future perspectives in this promising area are inspired. KEY FINDINGS Nanoformulations have a promising potential in improving the physico-chemical properties, pharmacokinetics, delivery efficiency, and selectivity of lung cancer therapeutics. The key challenges that encounter their clinical translation include their structural intricacy, high production cost, scale-up issues, and unclear toxicity profiles. The application of biodegradable platforms improves the biosafety of lung cancer-targeted nanomedicine. Moreover, the design of novel targeting strategies that apply a lower number of components can promote their industrial scalability and deliver them to the market at affordable prices. CONCLUSIONS Nanomedicines have opened up new possibilities for treating lung cancer. Focusing on tackling the challenges that hinder their clinical translation will promote the future of this area of endeavor.
Collapse
Affiliation(s)
- Mahmoud A Younis
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Mohammad A Alsogaihi
- Pharma D Student, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Imran Saleem
- Nanomedicine, Formulation & Delivery Research Group, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
35
|
Hao Y, Ji H, Gao L, Qu Z, Zhao Y, Chen J, Wang X, Ma X, Zhang G, Zhang T. Self-assembled carrier-free formulations based on medicinal and food active ingredients. Biomater Sci 2024; 12:6253-6273. [PMID: 39523875 DOI: 10.1039/d4bm00893f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The popularity of medicinal plants, which have a unique system and are mostly used in compound form for the prevention and treatment of a wide range of diseases, is growing worldwide. In recent years, with advances in chemical separation and structural analysis techniques, many of the major bioactive molecules of medicinal plants have been identified. However, the active ingredients in medicinal plants often possess chemical characteristics, including poor water solubility, stability and bioavailability, which limit their therapeutic applications. To address this problem, self-assembly of small molecules from medicinal food sources provides a new strategy. Driven by various types of acting forces, medicinal small molecules with modifiable groups, multiple sites of action, hydrophobic side chains, and rigid backbones with self-assembly properties are able to form various supramolecular network hydrogels, nanoparticles, micelles, and other self-assemblies. This review first summarizes the forms of self-assemblies such as supramolecular network hydrogels, nanoparticles, and micelles at the level of the action site, and discusses the recent studies on the active ingredients in medicinal plants that can be used for self-assembly, in addition to summarizing the advantages of self-assemblies for a variety of disease applications, including wound healing, antitumor, anticancer, and diabetes mellitus. Finally, the problems of self-assemblers and the possible directions for future development are presented. We firmly believe that self-assemblers have the potential to develop effective compounds from drug-food homologous plants, providing valuable information for drug research and new strategies and perspectives for the modernization of Chinese medicine.
Collapse
Affiliation(s)
- Yuan Hao
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Haixia Ji
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Li Gao
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Zhican Qu
- Shanxi Nanolattix Health Technology Co., Ltd, Taiyuan 030051, Shanxi, China
| | - Yinghu Zhao
- School of Environment and Safety Engineering, North University of China, Taiyuan 030051, Shanxi, China
| | - Jiahui Chen
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Xintao Wang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Xiaokai Ma
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Guangyu Zhang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| | - Taotao Zhang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, Shanxi, China.
| |
Collapse
|
36
|
Jia W, Li R, Zou F, Li M, Weng H, Shen Q, Qi G, Zhou R, Shi Y, Gu G, Wang F, Chen Z. Decorating Delivery Vehicles Using Hyaluronic Acid Oligosaccharides Enables Active Targeting Toward Cancer and Minimizes Adverse Effect of Chemotherapeutics. Adv Healthc Mater 2024; 13:e2402158. [PMID: 39221507 DOI: 10.1002/adhm.202402158] [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: 06/12/2024] [Revised: 08/19/2024] [Indexed: 09/04/2024]
Abstract
The major drawback of conventional chemotherapeutic treatment is the non-specificity or inability to ascertain and target cancerous cells directly. In this study, an active targeting strategy that is poised to carry the anticancer agents to the desired sites for therapeutic action while avoiding toxicity to normal organs is provided. The active targeting of delivery vehicles is achieved by ligand-receptor interactions, in particular the specific binding between hyaluronic acid oligosaccharides (oHAs) and CD44 receptors. This study first prepares oHAs by the size-exclusion chromatography and utilizes them to decorate chitosan (CTS) as basic materials (oHAs-CTS) for drug delivery, then fabricates oHAs-CTS into micro/nanoscale carriers to encapsulate agents for cancer chemotherapy. The oHAs-CTS micro/nanocarriers exhibit high drug encapsulation efficiency (58-87%), and the drug releases present a sustained behavior. Notably, oHAs-CTS delivery vehicles display an enhanced active targeting toward cancers and alleviate the cytotoxic effects on normal cells. Additionally, in vivo results show that drug-laden oHAs-CTS nanocarriers demonstrate a significant inhibitory effect on 4 T1 tumors without any toxicity to the major organs. Taken together, the findings highlight the potential of oHAs-CTS micro/nanospheres as delivery vehicles with enhanced active targeted capability toward cancers and minimized adverse effects of chemotherapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Weibin Jia
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong, SAR, 999077, China
| | - Runrun Li
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Fengjuan Zou
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Min Li
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Hongjuan Weng
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Qianqian Shen
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Guozhen Qi
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Ruipiao Zhou
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Yikang Shi
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Guofeng Gu
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| | - Fengshan Wang
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
- School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, China
| | - Zonggang Chen
- National Glycoengineering Research Center and Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Shandong University, Qingdao, 266237, China
| |
Collapse
|
37
|
Puvvada N, Shaik MAS, Samanta D, Shaw M, Mondal I, Basu R, Bhattacharya A, Pathak A. Biocompatible fluorescent carbon nanoparticles as nanocarriers for targeted delivery of tamoxifen for regression of Breast carcinoma. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 321:124721. [PMID: 38943755 DOI: 10.1016/j.saa.2024.124721] [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: 02/27/2024] [Revised: 05/14/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
Breast cancer (BC) is the most common malignancy among females worldwide, and its high metastasis rates are the leading cause of death just after lung cancer. Currently, tamoxifen (TAM) is a hydrophobic anticancer agent and a selective estrogen modulator (SERM), approved by the FDA that has shown potential anticancer activity against BC, but the non-targeted delivery has serious side effects that limit its ubiquitous utility. Therefore, releasing anti-cancer drugs precisely to the tumor site can improve efficacy and reduce the side effects on the body. Nanotechnology has emerged as one of the most important strategies to solve the issue of overdose TAM toxicity, owing to the ability of nano-enabled formulations to deliver desirable quantity of TAM to cancer cells over a longer period of time. In view of this, use of fluorescent carbon nanoparticles in targeted drug delivery holds novel promise for improving the efficacy, safety, and specificity of TAM therapy. Here, we synthesized biocompatible carbon nanoparticles (CNPs) using chitosan molecules without any toxic surface passivating agent. Synthesized CNPs exhibit good water dispersibility and emit intense blue fluorescence upon excitation (360 nm source). The surface of the CNPs has been functionalized with folate using click chemistry to improve the targeted drug uptake by the malignant cell. The pH difference between cancer and normal cells was successfully exploited to trigger TAM release at the target site. After six hours of incubation, CNPs released ∼ 74 % of the TAM drug in acidic pH. In vitro, studies have also demonstrated that after treatment with the synthesized CNPs, significant inhibition of the tumor growth could be achieved.
Collapse
Affiliation(s)
- Nagaprasad Puvvada
- Department of Chemistry, Indian Institute of Technology Kharagpur, West Bengal 721302, India; Department of Chemistry, School of Advanced Sciences, VIT-AP University, Vijayawada, Andhra Pradesh 522237, India
| | - Md Abdus Salam Shaik
- Department of Chemistry, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Dipanjan Samanta
- Department of Chemistry, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Manisha Shaw
- Department of Chemistry, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Imran Mondal
- Department of Chemistry, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Rajarshi Basu
- Department of Chemistry, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Angana Bhattacharya
- Department of Chemistry, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Amita Pathak
- Department of Chemistry, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
38
|
Babu LT, Roy N, Dasgupta T, Ghosh S, Tamizhselvi R, Paira P. Engineering biotin anchored-MWCNTs as a superb carrier for facile delivery of the potent Ru(II)-N^N scaffold in breast cancer cells. Chem Commun (Camb) 2024; 60:13376-13379. [PMID: 39324771 DOI: 10.1039/d4cc04276j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Ru(II)-complexes have been recognised as promising in treating cancer. However, targeted delivery is an important facet to augment the efficiency of drugs. Consequently, this article portrays the construction of biotinylated-MWCNTs as an SMVT-guided nano-platform for the precise delivery of our previously-developed potent Ru(II)-scaffold, making it more effective against MCF-7 cells.
Collapse
Affiliation(s)
- Lavanya Thilak Babu
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India.
| | - Nilmadhab Roy
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India.
| | - Tiasha Dasgupta
- Department of Bioscience, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Sreejani Ghosh
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India.
| | - Ramasamy Tamizhselvi
- Department of Bioscience, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India
| | - Priyankar Paira
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, Tamilnadu 632014, India.
| |
Collapse
|
39
|
Alghamdi WA, Alterary SS, Alarifi A, Ramu R, Khan MS, Afzal M. Exploring the interaction of curcumin with β-cyclodextrin and its binding with DNA: A combined spectroscopic and molecular docking study. Int J Biol Macromol 2024; 282:137238. [PMID: 39500426 DOI: 10.1016/j.ijbiomac.2024.137238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
At present, a major effort in biophysical studies has been paid towards exploring the interactions and release of therapeutic payloads to the specific site leaving behind healthy cells unaffected and hence, lower the drug-induced toxicity. For the purpose, interaction of β-bound CUR with calf thymus DNA (ctDNA) has been examined intensely using a series of biophysical methods like absorption, steady state fluorescence emission, and circular dichroism together with molecular docking study. The experimental analysis divulge that CUR interacts with both β-CD (although with different molar ratio) and DNA. However, the binding affinity of CUR with the target (DNA) is higher than it does with the β-CD. When β-CD-carried (10 mM) CUR (μM) (inclusion complex) comes near DNA (15-372 μM), CUR gets out from β-CD's void and approaches to binds with the DNA. The relocation of the probe occurred due to competitive binding of the CUR between β-CD and the DNA. The present investigation may provide a simple yet probable route for the transfer of encapsulated therapeutic payload of β-CD to the most relevant biomolecular target DNA.
Collapse
Affiliation(s)
- Waad A Alghamdi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Seham S Alterary
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah Alarifi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ramith Ramu
- Department of Biotechnology & Bioinformatics School of Life Science, JSS Academy of Higher Education & Research (Deemed to be University) Sri Shivarathreeshwara Nagara, Mysuru, Karnataka 570015, India
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohd Afzal
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
40
|
Ye Q, Zhang M, Li S, Liu W, Xu C, Li Y, Xie R. Controlled Stimulus-Responsive Delivery Systems for Osteoarthritis Treatment. Int J Mol Sci 2024; 25:11799. [PMID: 39519350 PMCID: PMC11545989 DOI: 10.3390/ijms252111799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA), a common and disabling degenerative joint disease, affects millions of people worldwide and imposes a considerable burden on patients and society due to its high prevalence and economic costs. The pathogenesis of OA is closely related to the progressive degradation of articular cartilage and the accompany inflammation; however, articular cartilage itself cannot heal and modulate the inflammation due to the lack of nerves, blood vessels, and lymph-vessels. Therefore, reliable and effective methods to treat OA remain highly desired. Local administration of drugs or bioactive materials by intra-articular injection of the delivery system represents a promising approach to treat OA, especially considering the prolonged joint retention, cartilage or chondrocytes targeting, and stimuli-responsive release to achieve precision OA therapy. This article summarizes and discusses the advances in the currently used delivery systems (nanoparticle, hydrogel, liposome, and microsphere) and then focuses on their applications in OA treatment from the perspective of endogenous stimulus (redox reactions, pH, enzymes, and temperature) and exogenous stimulus (near-infrared, magnetic, and ultrasound)-responsive release. Finally, the challenges and potential future directions for the development of nano-delivery systems are summarized.
Collapse
Affiliation(s)
- Qianwen Ye
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Mingshuo Zhang
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Shuyue Li
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Wenyue Liu
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Chunming Xu
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yumei Li
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Renjian Xie
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (Q.Y.); (M.Z.); (S.L.); (W.L.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
41
|
Yao Y, Zhou W, Cai K, Wen J, Zhang X. Advances in the study of the biological activity of polysaccharide-based carbon dots: A review. Int J Biol Macromol 2024; 281:135774. [PMID: 39419681 DOI: 10.1016/j.ijbiomac.2024.135774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024]
Abstract
Carbon dots have attracted worldwide interest due to their customizable nature, luminescent properties, and exceptional biocompatibility. In particular, biomass-derived carbon dots have attracted attention for their environmentally friendly and cost-effective synthesis. Recent research looks into how polysaccharides can be used to make carbon dots. Using them as starting materials for nanomaterials has benefits in terms of structure, morphology, and doping elements. Although research has extensively examined the optical properties of carbon dots, their potential biological applications have not been thoroughly investigated. This review mainly summarises the cytotoxicity and biological functions of polysaccharide-based carbon dots (e.g. agar, alginate, cellulose, carrageenan, chitosan, chitosan, starch, gelatin, etc.), such as antioxidant, antibacterial and anti-tumor functions, highlighting the different scenarios of the methods of preparation of carbon dots. The applications of carbon dots in food, biomedical sciences, soil fertilization, and power generation are highlighted by reviewing the low toxicity of carbon dots with safety and biocompatibility in human contact. Finally, the importance and challenges of polysaccharide-based carbon dots and the prospects and research directions of polysaccharide-based carbon dots are explained by comparing them with other nanomaterials.
Collapse
Affiliation(s)
- Yihuan Yao
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Wenzhao Zhou
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Kaiyue Cai
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaying Wen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xianfei Zhang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
42
|
Bhirud D, Bhattacharya S, Prajapati BG. Bioengineered carbohydrate polymers for colon-specific drug release: Current trends and future prospects. J Biomed Mater Res A 2024; 112:1860-1872. [PMID: 38721841 DOI: 10.1002/jbm.a.37732] [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: 03/10/2024] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 09/03/2024]
Abstract
The worldwide health burden of colorectal cancer is still substantial, and traditional chemotherapeutic drugs sometimes have poor selectivity, which can result in systemic toxicity and unfavorable side effects. For colon-specific medication delivery, bioengineered carbohydrate polymers have shown promise as carriers. They may enhance treatment effectiveness while minimizing systemic exposure and associated side effects. The unique properties of these manufactured or naturally occurring biopolymers, such as hyaluronic acid, chitosan, alginate, and pectin, enable targeted medicine release. These qualities can be changed to meet the physiological needs of the colon. In the context of colorectal cancer therapy, this article provides a comprehensive overview of current developments and prospective future directions in the field of bioengineered carbohydrate polymer synthesis for colon-specific drug delivery. We discuss numerous techniques for achieving colon-targeted drug release, including enzyme-sensitive polymers, pH-responsive devices, and microbiota-activated processes. To increase tumor selectivity and cellular uptake, we also examine the inclusion of active targeting approaches, such as conjugating specific ligands. Furthermore, we discuss the potential of combination treatment strategies, which use the coadministration of numerous therapeutic medications to target multiple pathways implicated in cancer growth and address drug resistance mechanisms. We address recent biomimetic advances that potentially improve the biocompatibility, cellular uptake, and tumor penetration of carbohydrate polymer-based nanocarriers. These methods involve protein corona engineering and cell membrane coating. Furthermore, we look at the possibility of intelligent and sensitive systems that may adjust their behaviors in response to certain inputs or feedback loops, allowing for precise and regulated drug distribution.
Collapse
Affiliation(s)
- Darshan Bhirud
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's NMIMS Deemed-to-be University, Shirpur, Maharashtra, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Shree S.K. Patel College of Pharmaceutical Education & Research, Mehsana, Gujarat, India
| |
Collapse
|
43
|
Merlin JPJ, Crous A, Abrahamse H. Combining Photodynamic Therapy and Targeted Drug Delivery Systems: Enhancing Mitochondrial Toxicity for Improved Cancer Outcomes. Int J Mol Sci 2024; 25:10796. [PMID: 39409125 PMCID: PMC11477455 DOI: 10.3390/ijms251910796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer treatment continues to be a substantial problem due to tumor complexities and persistence, demanding novel therapeutic techniques. This review investigates the synergistic potential of combining photodynamic therapy (PDT) and tailored medication delivery technologies to increase mitochondrial toxicity and improve cancer outcomes. PDT induces selective cellular damage and death by activating photosensitizers (PS) with certain wavelengths of light. However, PDT's efficacy can be hampered by issues such as poor light penetration and a lack of selectivity. To overcome these challenges, targeted drug delivery systems have emerged as a promising technique for precisely delivering therapeutic medicines to tumor cells while avoiding off-target effects. We investigate how these technologies can improve mitochondrial targeting and damage, which is critical for causing cancer cell death. The combination method seeks to capitalize on the advantages of both modalities: selective PDT activation and specific targeted drug delivery. We review current preclinical and clinical evidence supporting the efficacy of this combination therapy, focusing on case studies and experimental models. This review also addresses issues such as safety, distribution efficiency, resistance mechanisms, and costs. The prospects of further research include advances in photodynamic agents and medication delivery technology, with a focus on personalized treatment. In conclusion, combining PDT with targeted drug delivery systems provides a promising frontier in cancer therapy, with the ability to overcome current treatment limits and open the way for more effective, personalized cancer treatments.
Collapse
Affiliation(s)
- J. P. Jose Merlin
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa; (A.C.); (H.A.)
| | | | | |
Collapse
|
44
|
Kunjiappan S, Panneerselvam T, Pavadai P, Balakrishnan V, Pandian SRK, Palanisamy P, Sankaranarayanan M, Kabilan SJ, Sundaram GA, Tseng WL, Kumar ASK. Fabrication of folic acid-conjugated pyrimidine-2(5H)-thione-encapsulated curdlan gum-PEGamine nanoparticles for folate receptor targeting breast cancer cells. Int J Biol Macromol 2024; 277:134406. [PMID: 39097067 DOI: 10.1016/j.ijbiomac.2024.134406] [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: 02/19/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
In this study 5-((2-((3-methoxy benzylidene)-amino)-phenyl)-diazenyl)-4,6-diphenyl pyrimidine-2(5H)-thione was synthesized. The pharmacological applications of pyrimidine analogs are restricted due to their poor pharmacokinetic properties. As a solution, a microbial exopolysaccharide (curdlan gum) was used to synthesize folic acid-conjugated pyrimidine-2(5H)-thione-encapsulated curdlan gum-PEGamine nanoparticles (FA-Py-CG-PEGamine NPs). The results of physicochemical properties revealed that the fabricated FA-Py-CG-PEGamine NPs were between 100 and 400 nm in size with a majorly spherical shaped, crystalline nature, and the encapsulation efficiency and loading capacity were 79.04 ± 0.79 %, and 8.12 ± 0.39 % respectively. The drug release rate was significantly higher at pH 5.4 (80.14 ± 0.79 %) compared to pH 7.2. The cytotoxic potential of FA-Py-CG-PEGamine NPs against MCF-7 cells potentially reduced the number of cells after 24 h with 42.27 μg × mL-1 as IC50 value. The higher intracellular accumulation of pyrimidine-2(5H)-thione in MCF-7 cells leads to apoptosis, observed by AO/EBr staining and flow cytometry analysis. The highest pyrimidine-2(5H)-thione internalization in MCF-7 cells may be due to folate conjugated on the surface of curdlan gum nanoparticles. Further, internalized pyrimidine-2(5H)-thione increases the intracellular ROS level, leading to apoptosis and inducing the decalin in mitochondrial membrane potential. These outcomes demonstrated that the FA-Py-CG-PEGamine NPs were specificity-targeting folate receptors on the plasma membranes of MCF-7 Cells.
Collapse
Affiliation(s)
- Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamilnadu, India.
| | - Theivendren Panneerselvam
- Department of Pharmaceutical Chemistry, Swamy Vivekanandha College of Pharmacy, Elayampalayam, Namakkal 637205, Tamilnadu, India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, Bengaluru 560054, Karnataka, India
| | - Vanavil Balakrishnan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamilnadu, India
| | - Sureshbabu Ram Kumar Pandian
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamilnadu, India
| | - Ponnusamy Palanisamy
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India
| | - Murugesan Sankaranarayanan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani-333031, Rajasthan, India
| | | | - Ganeshraja Ayyakannu Sundaram
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Poonamallee High Road, Chennai 600 077, Tamilnadu, India
| | - Wei-Lung Tseng
- Department of Chemistry, National Sun Yat-sen University, No. 70, Lien-hai Road, Gushan District, Kaohsiung city 80424, Taiwan; School of Pharmacy, Kaohsiung Medical University, No. 100, Shiquan 1st Road, Sanmin District, Kaohsiung city 80708, Taiwan
| | | |
Collapse
|
45
|
Liu Y, Yang Z, Feng L, Xia Y, Wei G, Lu W. Advance in Nanomedicine for Improving Mucosal Penetration and Effective Therapy of Cervical Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2303772. [PMID: 37340569 DOI: 10.1002/smll.202303772] [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: 05/05/2023] [Indexed: 06/22/2023]
Abstract
Insufficient intratumor drug distribution and serious adverse effects are often associated with systemic chemotherapy for cervical cancer. Considering the location of cervical cancer, access to the cervix through the vagina may provide an alternative administration route for high drug amounts at the tumor site, minimal systemic exposure as well as convenience of non-invasive self-medication. Enormous progress has been made in nanomedicine to improve mucosal penetration and enhance the effectiveness of therapy for cervical cancer. This review article first introduce the physiological state of cervicovaginal cavity and the characteristics of intravaginal environment in cervical cancers. Based on introduction to the physiological state of cervicovaginal cavity and the characteristics of intravaginal environment in cervical cancers, both "first mucus-adhering then mucosal penetration" and "first mucus-penetrating then mucosal penetration" strategies are discussed with respect to mechanism, application condition, and examples. Finally, existing challenges and future directions are envisioned in the rational design, facile synthesis, and comprehensive utilization of nanomedicine for local therapy of cervical cancer. This review is expected to provide useful reference information for future research on nanomedicine for intravaginally administered formulations for topical treatment of cervical cancer.
Collapse
Affiliation(s)
- Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Ziyi Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Linglin Feng
- Shanghai Institute of Planned Parenthood Research, Key Laboratory of Contraceptives and Devices Research (NPFPC), Shanghai Engineer and Technology Research Center of Reproductive Health Drug and Devices, Shanghai, 200032, China
| | - Yu Xia
- Yangtze River Pharmaceutical Group Co., Ltd., Taizhou, Jiangsu, 225300, China
| | - Gang Wei
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| |
Collapse
|
46
|
Ara N, Hafeez A, Kushwaha SP. Repurposing simvastatin in cancer treatment: an updated review on pharmacological and nanotechnological aspects. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7377-7393. [PMID: 38748226 DOI: 10.1007/s00210-024-03151-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/06/2024] [Indexed: 10/04/2024]
Abstract
Management of cancer is challenging due to non-targeting and high side effect issues. Drug repurposing is an innovative method for employing medications for other disease therapy in addition to their original use. Simvastatin, a 3-hydroxy-3-methylglutaryl coenzyme-A reductase inhibitor, is a lipid-lowering drug that is being studied for the treatment of cancer in various in vitro and in vivo models. Nanotechnology offers a potential platform for incorporation of drugs with enhanced pharmaceutical (solubility, release characteristics, stability, etc.) and biological characteristics (targeting, pharmacokinetic, pharmacodynamic). Utilizing a variety of resources such as Scopus, Springer, Web of Science, Elsevier, Bentham Science, Taylor & Francis, and PubMed, a thorough literature search was carried out by looking through electronic records published between 2003 and 2024. The keywords used were simvastatin, drug repurposing, anti-cancer simvastatin, pharmaceutical properties of simvastatin, simvastatin nanoformulations, simvastatin patents, clinical trials, etc. Numerous articles were looked for, filtered, checked out, and incorporated. Pure simvastatin has been researched as a repurposed medication for the treatment of cancer in several in vitro and in vivo models, such as carcinoma of the lung, colon, liver, prostate, breast, and skin. Simvastatin also incorporated into different nanocarriers (nanosuspensions, microparticles/nanoparticles, liposomes, and nanostructured lipid carriers) and showed improvement in solubility, bioavailability, drug loading, release kinetics, and targeting. Clinical trial and patent reports suggest potential of simvastatin in cancer therapy. The preclinical studies of pure simvastatin in in vitro and in vivo models showed the potential for its ability to inhibit cancer cell growth and further incorporation into nanoformulations strengthened its preclinical and pharmaceutical characteristics.
Collapse
Affiliation(s)
- Nargis Ara
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Abdul Hafeez
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Shom Prakash Kushwaha
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
47
|
Nakka NMR, Rachamala HK, Angom RS, Indla NR, Dutta SK, Wang E, Bhattacharya S, Sesha Sainath AV, Babiker H, Pal K, Mukhopadhyay D. Dual drug-loaded tumor-targeted polymeric nanoparticles for enhancing therapeutic response in pancreatic ductal adenocarcinoma. Mater Today Bio 2024; 28:101199. [PMID: 39205875 PMCID: PMC11357805 DOI: 10.1016/j.mtbio.2024.101199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease where standard-of-care chemotherapeutic drugs have limited efficacy due to the development of drug resistance and poor drug delivery caused by a highly desmoplastic tumor microenvironment. Combining multiple drugs in a tumor-targeting carrier would be a favorable approach to overcome these limitations. Hence, a tumor-targeted peptide (TTP) conjugated amphiphilic tri-block copolymer was developed to make targeted polymer nanoparticles (TTP-PNPs) serving as a vehicle for carrying gemcitabine (Gem), paclitaxel (PTX), and their combination (Gem + PTX). The TTP-PNPs in the form of empty polymer (P), single drug-loaded [P(Gem) and P(PTX)], and dual drug-loaded [P(Gem + PTX)] polymer nanoformulations exhibited stable and homogenous spherical shapes with 110-160 nm size. These nanoformulations demonstrated excellent stability under in vitro physiological conditions and led to an efficient release of the drugs in the presence of reduced glutathione (GSH). The efficacy of these nanoparticles was thoroughly evaluated in vitro and in vivo, demonstrating a notable capacity to selectively target and restrict PDAC cells (PANC-1 and KPC) growth. The cellular uptake and biodistribution study showed a significantly higher tumor-targeting ability of TTP-PNPs than PNPs without TTP. Notably, P(Gem + PTX) exhibited the lowest IC50 compared to all other controls and showed heightened synergistic effects in both cell lines. Furthermore, P(Gem + PTX) showed a significantly better tumor reduction and median overall survival in mouse models than single drug-loaded TTP-PNPs or a combination of free drugs (Gem + PTX). In summary, our TTP-PNP system shows great promise as a novel platform for delivering Gem + PTX specifically to pancreatic cancer (PC), maximizing the therapeutic benefits with lower concentrations of the drugs and potentially reducing toxic side effects.
Collapse
Affiliation(s)
- Naga Malleswara Rao Nakka
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
| | - Hari Krishnareddy Rachamala
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
| | - Nagamalleswara Rao Indla
- Polymers and Functional Materials and Fluoro-Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
| | - Annadanam V. Sesha Sainath
- Polymers and Functional Materials and Fluoro-Agrochemicals Department, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Hani Babiker
- Department of Medicine, Division of Hematology-Oncology, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Sciences, Jacksonville, FL, 32224, USA
| |
Collapse
|
48
|
Hajinezhad MR, Roostaee M, Nikfarjam Z, Rastegar S, Sargazi G, Barani M, Sargazi S. Exploring the potential of silymarin-loaded nanovesicles as an effective drug delivery system for cancer therapy: in vivo, in vitro, and in silico experiments. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7017-7036. [PMID: 38630254 DOI: 10.1007/s00210-024-03099-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/11/2024] [Indexed: 09/25/2024]
Abstract
We aimed to perform a comprehensive study on the development and characterization of silymarin (Syl)-loaded niosomes as potential drug delivery systems. The results demonstrate significant novelty and promising outcomes in terms of morphology, size distribution, encapsulation efficiency, in vitro release behavior, free energy profiles of Syl across the niosome bilayer, hydrogen bonding interactions, antimicrobial properties, cytotoxicity, and in vivo evaluations. The physical appearance, size, and morphology assessment of free niosomes and Syl-loaded niosomes indicated stable and well-formed vesicular structures suitable for drug delivery. Transmission electron microscopy (TEM) analysis revealed spherical shapes with distinct sizes for each formulation, confirming uniform distribution. Dynamic light scattering (DLS) analysis confirmed the size distribution results with higher polydispersity index for Syl-loaded niosomes. The encapsulation efficiency of Syl in the niosomes was remarkable at approximately 91%, ensuring protection and controlled release of the drug. In vitro release studies showed a sustained release profile for Syl-loaded niosomes, enhancing therapeutic efficacy over time. Free energy profiles analysis identified energy barriers hindering Syl permeation through the niosome bilayer, emphasizing challenges in drug delivery system design. Hydrogen bonding interactions between Syl and niosome components contributed to energy barriers, impacting drug permeability. Antimicrobial assessments revealed significant differences in inhibitory effects against S. aureus and E. coli. Cytotoxicity evaluations demonstrated the superior tumor-killing potential of Syl-loaded niosomes compared to free Syl. In vivo studies indicated niosome formulations' safety profiles in terms of liver and kidney parameters compared to bulk Syl, showcasing potential for clinical applications. Overall, this research highlights the promising potential of Syl-loaded niosomes as effective drug delivery systems with enhanced stability, controlled release, and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Mohammad Reza Hajinezhad
- Basic Veterinary Science Department, Veterinary Faculty, University of Zabol, P. O. Box. 98613-35856, Zabol, Iran
| | - Maryam Roostaee
- Department of Chemistry, Faculty of Sciences, Vali-e-Asr University of Rafsanjan, Rafsanjan, Iran
| | - Zahra Nikfarjam
- Department of Physical & Computational Chemistry, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran
| | - Sanaz Rastegar
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, 7616913555, Iran
| | - Ghasem Sargazi
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, 7616913555, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
49
|
Rana A, Matiyani M, Negi PB, Tiwari H, Garwal K, Basak S, Sahoo NG. Polyvinylpyrrolidone‐functionalized graphene oxide as a nanocarrier for dual‐drug delivery of quercetin and curcumin against HeLa cancer cells. JOURNAL OF VINYL AND ADDITIVE TECHNOLOGY 2024; 30:1241-1253. [DOI: 10.1002/vnl.22115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/30/2024] [Indexed: 01/06/2025]
Abstract
AbstractThis study is to develop a nanocarrier based on polyvinylpyrrolidone (PVP)‐functionalized graphene oxide (GO–PVP), loaded with both curcumin (CUR) and quercetin (QSR), and then its performance compared with nanocarriers carrying the drugs separately. The study also aimed to investigate the cytotoxic effects of these nanocarriers on HeLa cancer cells. To achieve this, GO was synthesized using a modified version of Hummer's method and subsequently functionalized with PVP. Drug loading onto the GO and GO–PVP nanocarriers was achieved through hydrophobic interactions. Furthermore, the ability of the nanocarriers to accommodate a single drug or a combination of drugs was examined. In our study, combined system shows higher drug loading, that is, 28.1% of QSR and 24.34% of CUR onto GO–PVP–QSR–CUR nanocarrier in comparison to single drug nanocarrier systems GO–PVP–QSR and GO–PVP–CUR which loaded 22.5% of QSR and 18.73% of CUR, respectively. Notably, the synthesized nanocarrier exhibited a pH‐sensitive drug release pattern. These results collectively suggest that GO–PVP–CUR–QSR displayed significantly higher cytotoxicity against HeLa cancer cells compared to both single‐drug nanocarrier systems at the specified concentrations. In addition, future pre‐clinical and clinical studies to evaluate the safety and efficacy of GO–PVP–CUR–QSR for cancer treatment are strongly recommended.Highlights
Developed nanocarrier based on polyvinylpyrrolidone functionalized GO (GO–PVP).
The GO–PVP nanocarrier was loaded with both curcumin (CUR) and quercetin (QSR).
GO–PVP displays a higher loading capacity for both QSR and CUR compared to GO.
QSR‐ and CUR‐loaded GO–PVP nanocarriers exhibited higher cytotoxic effects.
Collapse
Affiliation(s)
- Anita Rana
- Prof. Rajendra Singh Nanoscience and Nanotechnology Centre, Department of Chemistry, D.S.B. Campus Kumaun University Nainital India
- Institute of Macromolecular Chemistry Academy of Science of the Czech Republic Prague 6 Czech Republic
| | - Monika Matiyani
- Prof. Rajendra Singh Nanoscience and Nanotechnology Centre, Department of Chemistry, D.S.B. Campus Kumaun University Nainital India
- Institute of Macromolecular Chemistry Academy of Science of the Czech Republic Prague 6 Czech Republic
| | - Pushpa Bhakuni Negi
- Department of Chemistry Graphic Era Hill University, Bhimtal Campus Nainital India
| | - Himani Tiwari
- Prof. Rajendra Singh Nanoscience and Nanotechnology Centre, Department of Chemistry, D.S.B. Campus Kumaun University Nainital India
| | - Kamal Garwal
- Prof. Rajendra Singh Nanoscience and Nanotechnology Centre, Department of Chemistry, D.S.B. Campus Kumaun University Nainital India
| | - Souvik Basak
- Department of Pharmaceutical Chemistry Dr. B.C. Roy College of Pharmacy & Allied Health Sciences Durgapur India
| | - Nanda Gopal Sahoo
- Prof. Rajendra Singh Nanoscience and Nanotechnology Centre, Department of Chemistry, D.S.B. Campus Kumaun University Nainital India
| |
Collapse
|
50
|
Ou X, Gao G, Habaz IA, Wang Y. Mechanisms of resistance to tyrosine kinase inhibitor-targeted therapy and overcoming strategies. MedComm (Beijing) 2024; 5:e694. [PMID: 39184861 PMCID: PMC11344283 DOI: 10.1002/mco2.694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
Tyrosine kinase inhibitor (TKI)-targeted therapy has revolutionized cancer treatment by selectively blocking specific signaling pathways crucial for tumor growth, offering improved outcomes with fewer side effects compared with conventional chemotherapy. However, despite their initial effectiveness, resistance to TKIs remains a significant challenge in clinical practice. Understanding the mechanisms underlying TKI resistance is paramount for improving patient outcomes and developing more effective treatment strategies. In this review, we explored various mechanisms contributing to TKI resistance, including on-target mechanisms and off-target mechanisms, as well as changes in the tumor histology and tumor microenvironment (intrinsic mechanisms). Additionally, we summarized current therapeutic approaches aiming at circumventing TKI resistance, including the development of next-generation TKIs and combination therapies. We also discussed emerging strategies such as the use of dual-targeted antibodies and PROteolysis Targeting Chimeras. Furthermore, we explored future directions in TKI-targeted therapy, including the methods for detecting and monitoring drug resistance during treatment, identification of novel targets, exploration of dual-acting kinase inhibitors, application of nanotechnologies in targeted therapy, and so on. Overall, this review provides a comprehensive overview of the challenges and opportunities in TKI-targeted therapy, aiming to advance our understanding of resistance mechanisms and guide the development of more effective therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Xuejin Ou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Ge Gao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China HospitalSichuan UniversityChengduChina
| | - Inbar A. Habaz
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|