1
|
Wei L, Wang H, Ye X, Yue J, Guo H, Mao D, Li X, Sun Y, Liu C, Liu Y, Chen Y. Oxymatrine and astragaloside IV co-loaded liposomes: Scale-up purposes and their enhancement of anti-PD-1 efficacy against breast cancer. Mater Today Bio 2025; 32:101634. [PMID: 40177381 PMCID: PMC11964553 DOI: 10.1016/j.mtbio.2025.101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 04/05/2025] Open
Abstract
The response rate of programmed cell death protein-1 (PD-1) inhibitors in breast cancer remains unsatisfactory, primarily due to the limited infiltration and activity of tumor-infiltrating T lymphocytes (TILs). Previous studies demonstrated that oxymatrine (Om) and astragaloside IV (As) could enhance TIL infiltration and function by inhibiting cancer-associated fibroblasts (CAFs) and promoting mitochondrial activity in TILs, respectively. Thus, combining Om and As may be a promising strategy to improve the antitumor effects of PD-1 inhibitors in breast cancer. However, co-delivery above drugs into breast cancer tissue is challenging due to their low bioavailability and distinct physicochemical properties. This study addresses this challenge by formulating Om and As co-loaded liposomes (Om-As-Lip) and comparing the scale-up production methods: high-pressure homogenization (EP-HPH) and microfluidics. Om-As-Lip prepared via microfluidics demonstrated superior entrapment efficiency (As: 99.03 ± 0.04 %, Om: 67.01 ± 0.02 %) and a significantly higher production rate (22.12 mL/min) compared to EP-HPH (1.19 mL/min). Additionally, Om-As-Lip produced by microfluidics increased the area under the curve (AUC) (Om: 6.17-fold, As: 2.07-fold) and maximum concentration (Cmax) (Om: 1.58-fold, As: 3.49-fold) compared to the free drugs. Importantly, Om-As-Lip enhanced the antitumor efficacy of α-PD-1 by inhibiting CAF activation and boosting TIL activity, resulting in a tumor inhibition rate of 61.2 % and extended survival in mice. This work presents a novel perspective for scaling up co-delivered formulations of drugs with differing polarities to improve breast cancer immunotherapy.
Collapse
Affiliation(s)
- Liangyin Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Hong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xietao Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Junfan Yue
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Hong Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Dengxuan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xia Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yeyang Sun
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Congyan Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Research Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
2
|
Liu L, Tu B, Sun Y, Liao L, Lu X, Liu E, Huang Y. Nanobody-based drug delivery systems for cancer therapy. J Control Release 2025; 381:113562. [PMID: 39993634 DOI: 10.1016/j.jconrel.2025.02.058] [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: 11/11/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Targeted delivery can elevate the local drug concentration within tumor tissues, while minimizing drug distribution to normal tissues, thus enhancing the effectiveness of anti-tumor medications and reducing adverse effects and systemic toxicities. Nanobodies, the novel molecular pattern of antibodies characterized by their small size, high stability, strong specificity, and low immunogenicity, have been extensively applied in targeted drug delivery for tumor therapy. This review discusses structural disparities and functional advantages of nanobodies compared to other antibody fragments and full-length antibody. It also highlights nanobody applications in targeted tumor therapy, focusing on their use in modifying delivery systems, e.g., liposomes, EVs, micelles, albumin nanoparticles, gold nanoparticles, polymeric nanoparticles, and as nanobody-drug conjugates. This review delves into the methods applied for integrating nanobodies into different drug delivery carriers, in order to provide useful information for researchers developing nanobody-based targeted drug delivery systems.
Collapse
Affiliation(s)
- Lin Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Bin Tu
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yao Sun
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Lingling Liao
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Xiaoling Lu
- College of Stomatology, Guangxi Medical University, Nanning 530021, China
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Yongzhuo Huang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| |
Collapse
|
3
|
Sun Z, Fu H, Zhang R, Wang H, Shen S, Zhao C, Li X, Sun Y, Li Y, Li Y. Advances in chemically modified HSA as a multifunctional carrier for transforming cancer therapy regimens. Int J Biol Macromol 2025; 305:141373. [PMID: 39988174 DOI: 10.1016/j.ijbiomac.2025.141373] [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: 12/11/2024] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Human serum albumin (HSA) is a versatile, biodegradable, biocompatible, non-toxic, and non-immunogenic protein nanocarrier, making it an ideal platform for developing advanced drug delivery systems. These properties have garnered significant attention in utilizing HSA nanoparticles for the safe and efficient delivery of chemotherapeutic agents. HSA-based nanoparticles can be surface-modified with various ligands to enable tumor-targeted drug delivery, enhancing therapeutic specificity and efficacy. Furthermore, the multifunctionality of HSA nanoparticles offers promising strategies to overcome challenges in cancer therapy, including poor bioavailability, off-target toxicity, and drug resistance. This review highlights the structural features of HSA, explores its diverse modifications to improve drug-binding affinity and targeting ability, and discusses its potential as a multifunctional carrier in oncology. By summarizing the latest advances in HSA modification techniques and applications, this review provides a comprehensive perspective on the future of protein-based drug delivery systems in tumor therapy.
Collapse
Affiliation(s)
- Zheng Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Hui Fu
- School of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruixuan Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Hui Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shiyang Shen
- School of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chengcheng Zhao
- Experimental Teaching and Practical Training Center, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiuyan Li
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yujiao Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yunfei Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yingpeng Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
4
|
Uti DE, Atangwho IJ, Alum EU, Ntaobeten E, Obeten UN, Bawa I, Agada SA, Ukam CIO, Egbung GE. Antioxidants in cancer therapy mitigating lipid peroxidation without compromising treatment through nanotechnology. DISCOVER NANO 2025; 20:70. [PMID: 40272665 DOI: 10.1186/s11671-025-04248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/27/2025]
Abstract
BACKGROUND Cancer treatments often exploit oxidative stress to selectively kill tumour cells by disrupting their lipid peroxidation membranes and inhibiting antioxidant enzymes. However, lipid peroxidation plays a dual role in cancer progression, acting as both a tumour promoter and a suppressor. Balancing oxidative stress through antioxidant therapy remains a challenge, as excessive antioxidant activity may compromise the efficacy of chemotherapy and radiotherapy. AIM This review explores the role of antioxidants in mitigating lipid peroxidation in cancer therapy while maintaining treatment efficacy. It highlights recent advancements in nanotechnology-based targeted antioxidant delivery to optimize therapeutic outcomes. METHODS A comprehensive literature review was conducted using reputable databases, including PubMed, Scopus, Web of Science, and ScienceDirect. The search focused on publications from the past five years (2020-2025), supplemented by relevant studies from earlier years. Keywords such as "antioxidants," "lipid peroxidation," "nanotechnology in cancer therapy," and "oxidative stress" were utilized. Relevant articles were critically analysed, and graphical illustrations were created. RESULTS Emerging evidence suggests that nanoparticles, including liposomes, polymeric nanoparticles, metal-organic frameworks, and others, can effectively encapsulate and control the release of antioxidants in tumour cells while minimizing systemic toxicity. Stimuli-responsive carriers with tumour-specific targeting mechanisms further enhance antioxidant delivery. Studies indicate that these strategies help preserve normal cells, mitigate oxidative stress-related damage, and improve treatment efficacy. However, challenges such as bioavailability, stability, and potential interactions with standard therapies remain. CONCLUSION Integrating nanotechnology with antioxidant-based interventions presents a promising approach for optimizing cancer therapy. Future research should focus on refining lipid peroxidation modulation strategies, assessing oxidative stress profiles during treatment, and employing biomarkers to determine optimal antioxidant dosing. A balanced approach to antioxidant use may enhance therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
- Daniel Ejim Uti
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria.
| | - Item Justin Atangwho
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Esther Ugo Alum
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda
| | - Emmanuella Ntaobeten
- Department of Cancer and Haematology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Uket Nta Obeten
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| | - Inalegwu Bawa
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | - Samuel A Agada
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | | | - Godwin Eneji Egbung
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
5
|
Jin H, Noh W, Kyung K, Yeo WS, Song YH, Heo YS, Kim DE. Aptamer- vs Fab-Conjugated Liposomes: A Comparative Study in Targeting Acute Myeloid Leukemia Cells. Bioconjug Chem 2025; 36:815-822. [PMID: 40148126 DOI: 10.1021/acs.bioconjchem.5c00065] [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: 03/29/2025]
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy characterized by uncontrolled proliferation of abnormal myeloid cells with a generally poor prognosis despite advancements in chemotherapy and stem cell transplantation. To enhance therapeutic efficacy and minimize systemic toxicity, we designed liposomal nanoparticles functionalized with two distinct targeting ligands, a DNA aptamer or fragment-antigen-binding (Fab) antibody, targeting the surface marker transmembrane glycoprotein CD33 antigen (CD33) on AML cells. Aptamer- and Fab-conjugated liposomes (Apt-Lipm and Fab-Lipm, respectively) were prepared and tested for cellular uptake by CD33-positive AML cell lines. Comparative studies revealed that Fab-Lipm exhibited significantly superior binding affinity, targeting efficiency, and cellular uptake compared with Apt-Lipm. Furthermore, we demonstrated the intracellular distribution and endocytic pathways of Fab-Lipm during the cellular uptake. This comparative study of aptamer- and Fab-conjugated liposomes suggests that the Fab-conjugated liposomal system offers enhanced precision in targeting AML cells for the development of effective therapeutic strategies against hematologic malignancies.
Collapse
Affiliation(s)
- Hyesoo Jin
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Wooseong Noh
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Kangwuk Kyung
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Woon-Seok Yeo
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Ye Han Song
- Department of Chemistry, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Yong-Seok Heo
- Department of Chemistry, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
- Uniwon PharmGene Inc, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
6
|
Hu J, Arvejeh PM, Bone S, Hett E, Marincola FM, Roh KH. Nanocarriers for cutting-edge cancer immunotherapies. J Transl Med 2025; 23:447. [PMID: 40234928 PMCID: PMC12001629 DOI: 10.1186/s12967-025-06435-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Cancer immunotherapy aims to harness the body's own immune system for effective and long-lasting elimination of malignant neoplastic tissues. Owing to the advance in understanding of cancer pathology and immunology, many novel strategies for enhancing immunological responses against various cancers have been successfully developed, and some have translated into excellent clinical outcomes. As one promising strategy for the next generation of immunotherapies, activating the multi-cellular network (MCN) within the tumor microenvironment (TME) to deploy multiple mechanisms of action (MOAs) has attracted significant attention. To achieve this effectively and safely, delivering multiple or pleiotropic therapeutic cargoes to the targeted sites of cancerous tissues, cells, and intracellular organelles is critical, for which numerous nanocarriers have been developed and leveraged. In this review, we first introduce therapeutic payloads categorized according to their predicted functions in cancer immunotherapy and their physicochemical structures and forms. Then, various nanocarriers, along with their unique characteristics, properties, advantages, and limitations, are introduced with notable recent applications in cancer immunotherapy. Following discussions on targeting strategies, a summary of each nanocarrier matching with suitable therapeutic cargoes is provided with comprehensive background information for designing cancer immunotherapy regimens.
Collapse
Affiliation(s)
- Joyce Hu
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | - Pooria M Arvejeh
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sydney Bone
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | - Erik Hett
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | | | - Kyung-Ho Roh
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
- Biotechnology Science and Engineering Program, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
| |
Collapse
|
7
|
Jawad RAM, Mshimesh BAR, Al-Mayah QS, Al-Alloosh F. A Case Study on Complete Pathological Response in Advanced Rectal Cancer Patient with Oxaliplatin-based Chemotherapy without Cumulative Neurotoxicity. J Gastrointest Cancer 2025; 56:99. [PMID: 40240738 DOI: 10.1007/s12029-025-01227-7] [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: 04/06/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND The pathological response in rectal cancer treatment provides insight into the molecular mechanisms, including genetic alterations and signaling pathways that influence tumor behavior and resistance to treatment. CASE PRESENTATION This report describes a 34-year-old Iraqi male diagnosed with stage III rectal cancer who achieved a complete pathological response following treatment with oxaliplatin-based chemotherapy. Notably, this outcome was achieved without the administration of chemoradiotherapy or the occurrence of neurotoxicity despite the efficacious cumulative‑dose administration (1700 mg/m2) of oxaliplatin. Genomic analysis revealed the presence of a heterozygous (Ile/Val) genotype in the GSTP1 gene, which may have contributed to the observed treatment response. CONCLUSIONS Genetic biomarkers play a crucial role in refining treatment strategies by enabling a more precise selection of patients who may safely forgo radiotherapy, thereby minimizing its associated toxicities. Additionally, molecular profiling can help predict susceptibility to oxaliplatin-induced neurotoxicity, facilitating dose adjustments or alternative therapeutic approaches to enhance treatment tolerance and long-term quality of life. Our findings highlight the importance of molecular profiling in optimizing treatment strategies while minimizing toxicity, especially in situations where radiological assessments suggest residual disease or produce unclear results.
Collapse
Affiliation(s)
- Rehab A M Jawad
- Department of Pharmacology and Toxicology, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq.
- Ministry of Health, Kimadia, Baghdad, Iraq.
| | | | - Qasim S Al-Mayah
- Medical Research Unit, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | | |
Collapse
|
8
|
Peng L, Gao Z, Liang Y, Guo X, Zhang Q, Cui D. Nanoparticle-based drug delivery systems: opportunities and challenges in the treatment of esophageal squamous cell carcinoma (ESCC). NANOSCALE 2025; 17:8270-8288. [PMID: 40052671 DOI: 10.1039/d4nr05114a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is an aggressive malignancy characterized by limited treatment options and poor prognosis. Nanoparticle-based drug delivery systems have emerged as a promising strategy to enhance cancer therapy efficacy by improving drug targeting, reducing toxicity, and enabling multifunctional applications. This review highlights some key types of nanoparticles, including liposomes, polymeric nanoparticles, metallic nanoparticles, dendrimers, and quantum dots, which could effectively improve the delivery of various drugs used in chemotherapy, radiotherapy, and immunotherapy, offering more precise and effective treatment options. With the ability to improve drug stability and overcome biological barriers, nanoparticle-based systems represent a transformative strategy for ESCC treatment. Despite some challenges, such as biocompatibility and scalability, the future of nanoparticle-based drug delivery holds great promise, particularly in the development of personalized nanomedicine and novel therapeutic approaches targeting the tumor microenvironment. With ongoing advancements, nanoparticle-based drug delivery systems hold immense potential to revolutionize ESCC treatment and improve patient outcomes.
Collapse
Affiliation(s)
- Linjia Peng
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Zixuan Gao
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Yanfeng Liang
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Xiaonan Guo
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Qiuli Zhang
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Daxiang Cui
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| |
Collapse
|
9
|
Radziunas-Salinas Y, Domínguez-Arca V, Pardo A, Cambón A, Taboada P, Prieto G. Long-chain cationic gemini surfactants as drug retention adjuvant on liposomes. A methodological approach with atorvastatin. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184419. [PMID: 40187473 DOI: 10.1016/j.bbamem.2025.184419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/14/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
This study delves into the development and characterization of dipalmitoyl phosphatidylcholine (DPPC) liposomes incorporated with gemini surfactant (tetradecamethylene-1,14 bis(dimethyl tetradecyl ammonium bromide); 14-14-14) and atorvastatin, aimed at enhancing drug delivery efficiency for cardiovascular diseases. The integration of gemini surfactants into liposomes is investigated for its potential to improve atorvastatin encapsulation and retention, addressing the drug's poor water solubility and the limitations of conventional liposomal systems. Through a combination of dynamic light scattering (DLS), differential scanning calorimetry (DSC), and molecular dynamics (MD) simulations, the study reveals that the presence of gemini surfactants significantly reduces liposome size and polydispersity, indicative of a more uniform and potentially unilamellar structure. DSC analysis highlights a decrease in transition temperatures and an alteration in transition symmetry, suggesting enhanced stability and a favourable drug release profile at physiological temperatures. MD simulations provide insight into the internalization mechanism of gemini surfactants and atorvastatin within the liposomal bilayer, demonstrating their mutual incorporation facilitated by polar interactions. Spectrophotometry-based retention studies further confirmed that liposomes containing gemini surfactants exhibit superior atorvastatin retention capabilities, nearly doubling the encapsulation efficiency compared to conventional liposomes. This research highlights the promising role of gemini surfactant-incorporated liposomes as an efficient drug delivery platform for cardiovascular therapeutics, offering insights into the molecular interactions and structural dynamics underlying their enhanced performance.
Collapse
Affiliation(s)
- Yago Radziunas-Salinas
- Applied Physics Department, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Vicente Domínguez-Arca
- Applied Physics Department, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Biosystems and Bioprocess Engineering (Bio2Eng) Group, Institute of Marine Research of Spanish Research Council, IIM-CSIC, 36208 Vigo, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Alberto Pardo
- Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Adriana Cambón
- Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Pablo Taboada
- Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Gerardo Prieto
- Applied Physics Department, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Colloids and Polymers Physics Group, Department of Physics of Particles, Faculty of Physics and Institute of Health Research (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
10
|
Shaikh S, Chary PS, Mehra NK. Tyrosine Kinase Inhibitor Lenvatinib Based Nano Formulations and Cutting-Edge Scale-Up Technologies in revolutionizing Cancer Therapy. ACS APPLIED BIO MATERIALS 2025; 8:1749-1784. [PMID: 40091597 DOI: 10.1021/acsabm.4c01527] [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: 03/19/2025]
Abstract
Lenvatinib (LEN), a tyrosine kinase inhibitor, has emerged as a promising therapeutic agent for various solid tumors. Nevertheless, a number of constraints, including diminished bioavailability, incapacity to elicit localized inflammation, and inability to selectively accumulate at the tumor site, may impede the comprehensive exploitation of its versatile tyrosine kinase inhibitory capabilities. In order to achieve targeted delivery of LEN while also reducing its high dose used in conventional therapeutics, nanoformulation approaches can be adopted. The integration of LEN into various nanoformulations, such as nanoparticles, nanocrystals, high density lipoproteins (HDLs), liposomes, and micelles, is discussed, highlighting the advantages of these innovative approaches in a comparative manner; however, given that the current methods of nanoformulation synthesis employ toxic organic solvents and chemicals, there is an imperative need for exploring alternative, environmentally friendly approaches. The multifaceted effects of nanocarriers have rendered them profoundly applicable within the biomedical domain, serving as instrumental entities in various capacities such as vehicles for drug delivery and genetic material, diagnostic agents, facilitators of photothermal therapy, and radiotherapy. However, the scalability of these nanotechnological methodologies must be rigorously investigated and addressed to refine drug delivery mechanisms. This endeavor offers promising prospects for revolutionizing strategies in cancer therapeutics, thereby laying the foundation for future research in scale-up techniques in the pursuit of more effective and less toxic therapies for cancer.
Collapse
Affiliation(s)
- Samia Shaikh
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| |
Collapse
|
11
|
Bao J, Li Z, Zhang D. β-elemene: A promising natural compound in lung cancer therapy. Eur J Pharmacol 2025; 997:177399. [PMID: 40064226 DOI: 10.1016/j.ejphar.2025.177399] [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: 12/04/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 04/18/2025]
Abstract
Lung cancer, a leading cause of cancer-related mortality globally, presents complex challenges in treatment and disease management. This review explores β-elemene, a sesquiterpene from Curcuma wenyujin, emphasising its pharmacological effects and therapeutic mechanisms in lung cancer. Focusing on its roles in modulating cellular pathways, this study details β-elemene's influence on apoptosis, autophagy, ferroptosis, hypoxic responses, metabolic shifts, and cell cycle arrest, as well as its impact on the tumour microenvironment and regulatory pathways (including PI3K/AKT, STAT3, AMPK/MAPK) and non-coding RNAs. The potential of β-elemene as a complementary agent in chemotherapy, radiotherapy, and hyperthermia therapy is examined, underscoring its capability to bolster treatment efficacy and counter drug resistance. The review also addresses current obstacles in clinical use, notably bioavailability issues, and explores innovative delivery systems like liposomes and microemulsions designed to enhance therapeutic delivery. Although preclinical studies indicate significant anti-tumor effects, further research is needed to address clinical translation challenges. Collectively, this review highlights β-elemene's multi-targeted therapeutic potential in lung cancer, advocating for ongoing research to refine its clinical use and optimize patient outcomes.
Collapse
Affiliation(s)
- Jiahui Bao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Zhiliang Li
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110001, China
| | - Dan Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| |
Collapse
|
12
|
Zhong X, Zhang Y, Wei J. Recent advances in ruthenium (III) complex-loaded nanomaterial for enhanced cancer therapy efficacy. Drug Dev Ind Pharm 2025; 51:169-179. [PMID: 39836522 DOI: 10.1080/03639045.2025.2455428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Amid the escalating global cancer incidence, the development of effective and safe anticancer drugs is a critical priority in medical research. Addressing the clinical shortcomings of ruthenium-based anticancer drugs are currently a prominent focus of research. SIGNIFICANCE AND METHODS Since the pioneering work with platinum derivatives, significant progress has been made in the fundamental studies of metal complexes for the treatment of a wide range of cancers, and there has been a growing interest in their properties and biomedical applications. Although chemotherapy is crucial in clinical tumor management, platinum(II) anticancer drugs like cisplatin and carboplatin suffer from severe toxicity and drug resistance issues. Among various metal-based drugs, ruthenium(III) complexes are notable for their selectivity, cytotoxic activity in vitro, and effective anticancer properties in vivo. Despite some drug candidates reaching late-stage clinical trials, their clinical application remains constrained by problems such as low solubility, poor stability, and inadequate cellular uptake. RESULTS The development of efficient and stable nanocarrier-based drug delivery systems for ruthenium(III) complexes, enhancing pharmacokinetic properties, and enabling slow, controlled release and targeted drug delivery, offers potential solutions to these limitations. CONCLUSIONS This review delves into the recent strides in nanomaterial-based drug delivery for ruthenium complexes, encompassing research on platinum (II) and ruthenium (III) metal complexes, nano-delivery system designs, and addresses pivotal challenges and future trajectories in this domain.
Collapse
Affiliation(s)
- Xuemei Zhong
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, China
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, China
| | - Jianhua Wei
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, China
| |
Collapse
|
13
|
Yang J, Shrestha A, Ramalingam L. Fishing for Solutions: How Pre-Conceptional Fish Oil Supplementation in Obese Fathers Reduces Risk of Non-Alcoholic Fatty Liver Disease in Offspring Mice. Mol Nutr Food Res 2025; 69:e202400452. [PMID: 39910853 PMCID: PMC11874265 DOI: 10.1002/mnfr.202400452] [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: 08/12/2024] [Revised: 11/30/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025]
Abstract
Metabolic dysfunction associated fatty liver disease (MAFLD) is a chronic condition with hepatic fat accumulation. The intergenerational effect of obesity has predominantly focused on mothers, with limited studies on paternal obesity. Nutritional intervention with fish oil (FO) has beneficial effects in reducing markers of obesity. We hypothesized that supplementing obese fathers with FO before conception could enhance the metabolic health of their offspring liver. Male mice were assigned to low-fat (LF), high fat (HF), or HF supplemented with FO for 10 weeks. Subsequently, these males were mated with females on a chow diet. Offspring were sacrificed at 8 weeks, and liver tissues were analyzed for gene expression and histology. Offspring body weight was not significantly impacted by paternal diet. However, male offspring of HF fathers had higher levels of markers of inflammation and fatty acid synthesis compared to offspring of LF fed fathers. Paternal FO supplementation significantly reduced fatty acid synthesis and glucose metabolism, while increasing fatty acid oxidation in male offspring, with a less pronounced effect in female offspring. These findings suggest that FO supplementation in obese fathers prior to conception attenuates the development of MAFLD in male offspring. This data underscores the significance of paternal nutritional intervention in promoting offspring health.
Collapse
Affiliation(s)
- Junhui Yang
- Department of Nutrition and Food StudiesSyracuse UniversitySyracuseNew YorkUSA
| | - Akriti Shrestha
- Department of Nutrition and Food StudiesSyracuse UniversitySyracuseNew YorkUSA
| | - Latha Ramalingam
- Department of Nutrition and Food StudiesSyracuse UniversitySyracuseNew YorkUSA
| |
Collapse
|
14
|
Loscertales E, Mateo J, España S. A comparative study of sensitizers and liposome composition in radiation-induced controlled drug release for cancer therapy. J Liposome Res 2025; 35:64-75. [PMID: 39258993 DOI: 10.1080/08982104.2024.2401800] [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: 05/08/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
This study investigates drug-loaded liposomes designed for controlled release under ionizing radiation to refine cancer treatment precision. Liposomes as carriers enable targeted chemotherapy delivery, reducing healthy tissue damage risk. Liposomes containing poly- or mono-unsaturated fatty acids and various sensitizing agents were assessed for responsiveness to UV light and γ photon irradiation including rose bengal (RB), protoporphyrin IX (PPIX), verteporfin (VP), cercosporin (CERC) and hypericin (HYP). Carboxyfluorescein (CF) was used as a surrogate for drug release measurements. VP and PPIX induced rapid drug release and lipid peroxidation under UV light, while RB prompted quick drug release under UV light and a modest immediate release under γ irradiation, eventually reaching full release a few hours after irradiation, demonstrating dose-dependent effects. Smaller liposomes displayed accelerated release, emphasizing size-dependent kinetics. In vitro analyses evaluated radiosensitizing effects of RB-loaded liposomes. Clonogenic assays indicated that RB-filled liposomes had minimal direct radiobiological effects but increased indirect radiation damage, as shown by the curvature of the cell survival curve. Our study sheds light on factors influencing liposomal drug release under ionizing radiation, spotlighting RB as a promising radiosensitizer requiring further investigation for cancer therapy potential.
Collapse
Affiliation(s)
- E Loscertales
- Grupo de Física Nuclear, EMFTEL & IPARCOS, Universidad Complutense de Madrid, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - J Mateo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - S España
- Grupo de Física Nuclear, EMFTEL & IPARCOS, Universidad Complutense de Madrid, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Institute for Physical and Information Technologies "Leonardo Torres Quevedo", ITEFI, Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
15
|
Bolideei M, Barzigar R, Gahrouei RB, Mohebbi E, Haider KH, Paul S, Paul MK, Mehran MJ. Applications of Gene Editing and Nanotechnology in Stem Cell-Based Therapies for Human Diseases. Stem Cell Rev Rep 2025:10.1007/s12015-025-10857-0. [PMID: 40014250 DOI: 10.1007/s12015-025-10857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Stem cell research is a dynamic and fast-advancing discipline with great promise for the treatment of diverse human disorders. The incorporation of gene editing technologies, including ZFNs, TALENs, and the CRISPR/Cas system, in conjunction with progress in nanotechnology, is fundamentally transforming stem cell therapy and research. These innovations not only provide a glimmer of optimism for patients and healthcare practitioners but also possess the capacity to radically reshape medical treatment paradigms. Gene editing and nanotechnology synergistically enhance stem cell-based therapies' precision, efficiency, and applicability, offering transformative potential for treating complex diseases and advancing regenerative medicine. Nevertheless, it is important to acknowledge that these technologies also give rise to ethical considerations and possible hazards, such as inadvertent genetic modifications and the development of genetically modified organisms, therefore creating a new age of designer infants. This review emphasizes the crucial significance of gene editing technologies and nanotechnology in the progress of stem cell treatments, particularly for degenerative pathologies and injuries. It emphasizes their capacity to restructure and comprehensively revolutionize medical treatment paradigms, providing fresh hope and optimism for patients and healthcare practitioners.
Collapse
Affiliation(s)
- Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Rambod Barzigar
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India
| | - Razieh Bahrami Gahrouei
- Department of Pharmacy PES College, Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka, India
| | - Elham Mohebbi
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois School of Medicine, Springfield, IL, USA
| | - Khawaja Husnain Haider
- Sulaiman AlRajhi Medical School, Al Bukayriyah, AlQaseem, 52726, Kingdom of Saudi Arabia
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA.
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Mohammad Javad Mehran
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India.
| |
Collapse
|
16
|
Bai J, Yang G, Yu Q, Chi Q, Zeng X, Qi W. SATB1 in cancer progression and metastasis: mechanisms and therapeutic potential. Front Oncol 2025; 15:1535929. [PMID: 40071088 PMCID: PMC11893431 DOI: 10.3389/fonc.2025.1535929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a major global health challenge, with prostate cancer, lung cancer, colorectal cancer, and breast cancer accounting for nearly half of all diagnoses. Despite advancements in cancer treatment, metastasis to distant organs continues to be the leading cause of cancer-related mortality. The progression of cancer involves the alteration of numerous genes, with dynamic changes in chromatin organization and histone modifications playing a critical role in regulating cancer-associated genes. Special AT-rich sequence-binding protein 1 (SATB1), a critical chromatin organizer, plays a pivotal role in cancer progression by regulating gene expression, chromatin remodeling, and cell signaling pathways. SATB1 binds to AT-rich DNA sequences, acting as a scaffold for chromatin-modifying enzymes and transcription factors, thus coordinating the regulation of extensive gene networks. Its overexpression has been implicated in a wide range of cancers and is associated with poor prognosis, aggressive tumor phenotypes, and enhanced epithelial-mesenchymal transition (EMT). Moreover, SATB1's activity is modulated by microRNAs (miRNAs) and post-translational modifications, further contributing to its complex regulatory functions. Given its crucial involvement in cancer progression and metastasis, SATB1 has emerged as a promising target for novel therapeutic strategies. This review delves into the molecular mechanisms of SATB1 in cancer and explores potential therapeutic approaches for targeting this key regulator in cancer treatment.
Collapse
Affiliation(s)
- Jinping Bai
- Department of Bioscience, Changchun Normal University, Changchun, China
| | - Gege Yang
- Department of Bioscience, Changchun Normal University, Changchun, China
| | - Qi Yu
- Department of Bioscience, Changchun Normal University, Changchun, China
| | - Qianya Chi
- Department of Bioscience, Changchun Normal University, Changchun, China
| | - Xianlu Zeng
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, China
| | - Wenjing Qi
- Department of Bioscience, Changchun Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, China
| |
Collapse
|
17
|
Sah SK, Ajay SA, Donadkar AD, Kamath AJ, Devan AR, Soman R, Kumar AR, Unni AR, Sithara MS, Sudheesh MS, Nath LR. Ternary complex of Kaempferol-Hydroxypropyl-β-Cyclodextrin-Liposomes against hepatocellular carcinoma: Preparation, validation, pharmacokinetics and efficacy studies. Int J Pharm 2025; 671:125261. [PMID: 39855283 DOI: 10.1016/j.ijpharm.2025.125261] [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: 11/19/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Kaempferol (KP), a GRAS-certified phytomolecule enrolled in Phase I trials, is reported with various biological effects including anticancer activity. However, its poor pharmacokinetic profile limits the translational utility. Studies indicate that liposomes incorporating cyclodextrin inclusion complexes improves the bioavailability of hydrophobic drugs. The present study focuses on preparing and validating a novel ternary complex of Kaempferol-Hydroxypropyl-β-Cyclodextrin-Liposomes (KP-HP-β-CD-Liposomes) that shows a particle size of 131.70 ± 0.10 nm, a zeta potential of -26.59 ± 0.42 mV, and a drug entrapment efficiency of 90.14 ± 0.25 %. The KP-HP-β-CD-Liposomes demonstrate stability under refrigerated conditions (2-8 °C) over a three-month period. Also, it doesn't exhibit any cytotoxicity in normal fibroblast cells even up to 48 mg/ml while it produces a dose dependent cytotoxicity in HepG2 cells. It shows a better cellular uptake in HepG2 cells in comparison with pure Kaempferol as evidenced by HPLC analysis. KP-HP-β-CD-Liposomes induce apoptosis in HepG2 cells as assessed by Acridine orange ethidium bromide staining. Pharmacokinetic studies on Sprague Dawley rats indicate a significant improvement in Cmax and AUC(0-∞) of Kaempferol. The tissue distribution studies show that KP-HP-β-CD-Liposomes are highly accumulated in liver. The KP-HP-β- CD-Liposomes inhibits the development of hepatic tumors in Syngeneic N1S1 animal models.
Collapse
Affiliation(s)
- Sunil Kumar Sah
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India; Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - S A Ajay
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India; Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - Asawari Dilip Donadkar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India; Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - Adithya Jayaprakash Kamath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India; Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - Aswathy R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India; Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - Rahul Soman
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - Ayana R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India; Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - Ashok R Unni
- Central Lab Animal Facility, Department of Veterinary Medicine, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - M S Sithara
- Central Lab Animal Facility, Department of Veterinary Medicine, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - M S Sudheesh
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041 Kerala, India.
| |
Collapse
|
18
|
Moni SS, Moshi JM, Matou-Nasri S, Alotaibi S, Hawsawi YM, Elmobark ME, Hakami AMS, Jeraiby MA, Sulayli AA, Moafa HN. Advances in Materials Science for Precision Melanoma Therapy: Nanotechnology-Enhanced Drug Delivery Systems. Pharmaceutics 2025; 17:296. [PMID: 40142960 PMCID: PMC11945159 DOI: 10.3390/pharmaceutics17030296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Melanoma, a highly aggressive form of skin cancer, poses a major therapeutic challenge due to its metastatic potential, resistance to conventional therapies, and the complexity of the tumor microenvironment (TME). Materials science and nanotechnology advances have led to using nanocarriers such as liposomes, dendrimers, polymeric nanoparticles, and metallic nanoparticles as transformative solutions for precision melanoma therapy. This review summarizes findings from Web of Science, PubMed, EMBASE, Scopus, and Google Scholar and highlights the role of nanotechnology in overcoming melanoma treatment barriers. Nanoparticles facilitate passive and active targeting through mechanisms such as the enhanced permeability and retention (EPR) effect and functionalization with tumor-specific ligands, thereby improving the accuracy of drug delivery and reducing systemic toxicity. Stimuli-responsive systems and multi-stage targeting further improve therapeutic precision and overcome challenges such as poor tumor penetration and drug resistance. Emerging therapeutic platforms combine diagnostic imaging with therapeutic delivery, paving the way for personalized medicine. However, there are still issues with scalability, biocompatibility, and regulatory compliance. This comprehensive review highlights the potential of integrating nanotechnology with advances in genetics and proteomics, scalable, and patient-specific therapies. These interdisciplinary innovations promise to redefine the treatment of melanoma and provide safer, more effective, and more accessible treatments. Continued research is essential to bridge the gap between evidence-based scientific advances and clinical applications.
Collapse
Affiliation(s)
- Sivakumar S. Moni
- College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Jobran M. Moshi
- Department of Medical Laboratory Technology, College of Nursing and Health Science, Jazan University, Jazan 45142, Saudi Arabia
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Sabine Matou-Nasri
- Blood and Cancer Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh 11481, Saudi Arabia;
- Biosciences Department, Faculty of the School for Systems Biology, George Mason University, Manassas, VA 22030, USA
| | - Shmoukh Alotaibi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah 23433, Saudi Arabia; (S.A.); (Y.M.H.)
| | - Yousef M. Hawsawi
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah 23433, Saudi Arabia; (S.A.); (Y.M.H.)
- Department of Biochemistry and Molecular Medicine, College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | | | | | - Mohammed A. Jeraiby
- Department of Basic Medical Science, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia;
| | - Ahmed A. Sulayli
- Laboratory Department, Prince Mohammed bin Nasser Hospital, Jazan Health Cluster, Jazan 82734, Saudi Arabia;
| | - Hassan N. Moafa
- Department of Public Health, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Quality and Patients Safety, Jazan University Hospital, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
19
|
Zhang W, Peng D, Cheng S, Ni R, Yang M, Cai Y, Chen J, Liu F, Liu Y. Inflammatory Cell-Targeted Delivery Systems for Myocardial Infarction Treatment. Bioengineering (Basel) 2025; 12:205. [PMID: 40001724 PMCID: PMC11852162 DOI: 10.3390/bioengineering12020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Myocardial infarction (MI) is a cardiovascular disease (CVD) with high morbidity and mortality worldwide, which is a serious threat to human life and health. Inflammatory and immune responses are initiated immediately after MI, and unbalanced inflammation post-MI can lead to cardiac dysfunction, scarring, and ventricular remodeling, emphasizing the critical need for an effective inflammation-regulating treatment. With the development of novel therapies, the drug delivery system specific to inflammatory cells offers significant potential. In this review, we introduce immune cells and fibroblasts involved in the development of MI and summarize the newly developed delivery systems related to the use of injectable hydrogels, cardiac patches, nanoparticles, and extracellular vesicles (EVs). Finally, we highlight the recent trends in the use of inflammatory cell-targeting drug delivery systems involving different strategies that facilitate the effective treatment of MI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yao Liu
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing 400042, China; (W.Z.); (D.P.)
| |
Collapse
|
20
|
Sebatana R, Kudzai KD, Magura A, Mdlophane A, Zeevaart JR, Sathekge M, Kahts M, Mdanda S, Witika BA. An Insight to Nanoliposomes as Smart Radiopharmaceutical Delivery Tools for Imaging Atherosclerotic Plaques: Positron Emission Tomography Applications. Pharmaceutics 2025; 17:240. [PMID: 40006607 PMCID: PMC11858949 DOI: 10.3390/pharmaceutics17020240] [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: 12/14/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Atherosclerosis is a chronic progressive disease which is known to cause acute cardiovascular events as well as cerebrovascular events with high mortality. Unlike many other diseases, atherosclerosis is often diagnosed only after an acute or fatal event. At present, the clinical problems of atherosclerosis mainly involve the difficulty in confirming the plaques or identifying the stability of the plaques in the early phase. In recent years, the development of nanotechnology has come with various advantages including non-invasive imaging enhancement, which can be studied for the imaging of atherosclerosis. For targeted imaging and atherosclerosis treatment, nanoliposomes provide enhanced stability, drug administration, extended circulation, and less toxicity. This review discusses the current advances in the development of tailored liposomal nano-radiopharmaceutical-based techniques and their applications to atherosclerotic plaque diagnosis. This review further highlights liposomal nano-radiopharmaceutical localisation and biodistribution-key processes in the pathophysiology of atherosclerosis. Finally, this review discusses the direction and future of liposomal nano-radiopharmaceuticals as a potential clinical tool for the assessment and diagnosis of atherosclerotic plaque.
Collapse
Affiliation(s)
- Reabetswe Sebatana
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
| | - Kahwenga D. Kudzai
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Allan Magura
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Amanda Mdlophane
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Jan Rijn Zeevaart
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
- Radiochemistry, The South African Nuclear Energy Corporation (Necsa) SOC Ltd., Pelindaba 0240, South Africa
| | - Mike Sathekge
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Maryke Kahts
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Sipho Mdanda
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Bwalya Angel Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| |
Collapse
|
21
|
Jiang T, Jia T, Yin Y, Li T, Song X, Feng W, Wang S, Ding L, Chen Y, Zhang Q. Cuproptosis-Inducing Functional Nanocomposites for Enhanced and Synergistic Cancer Radiotherapy. ACS NANO 2025; 19:5429-5446. [PMID: 39895200 DOI: 10.1021/acsnano.4c13753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Radiotherapy is crucial in local cancer management and needs advancements. Tumor cells elevate intracellular copper levels to promote growth and resist radiation; thus, targeted copper delivery to mitochondria could enhance radiotherapy by inducing cuproptosis in tumor cells. In this study, we engineered a multifunctional nanoliposome complex, termed Lipo-Ele@CuO2, which encapsulates both copper peroxide (CuO2) and the copper chelator elesclomol, which can delivery Cu ions to the mitochondria. The Lipo-Ele@CuO2 complex induces mitochondria-mediated cuproptosis in tumor cells and synergistically enhances the efficacy of radiotherapy. CuO2 acts as a copper donor and exhibits inherent sensitivity to acidic environments. Additionally, it depletes intracellular glutathione, thereby sensitizing cells to cuproptosis. Leveraging its pH-responsive properties in the acidic tumor microenvironment, the Lipo-Ele@CuO2 facilitate the controlled release of elesclomol, efficiently delivering copper ions to mitochondria at tumor sites. The combined in vitro and in vivo studies demonstrate that Lipo-Ele@CuO2-based therapy significantly improves antitumor efficacy and exhibits excellent safety profiles, effectively inducing cuproptosis in tumor cells and boosting the effectiveness of radiotherapy. Furthermore, metabolomic and transcriptomic analyses reveal that this combination therapy precipitates significant alterations in tumor energy metabolism, notably repressing genes related to iron-sulfur cluster assembly and glycolysis, thereby confirming the induction of cuproptosis. This therapeutic strategy provides a viable approach for addressing clinical radiotherapy resistance and demonstrates significant translational potential.
Collapse
Affiliation(s)
- Tiaoyan Jiang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China
| | - Tianying Jia
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China
| | - Yipengchen Yin
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China
| | - Tianyu Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Sheng Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Li Ding
- Department of Medical Ultrasound, National Clinical Research Center of Interventional Medicine, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Tongji University, Shanghai 200072, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang 325088, P. R. China
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, P. R. China
| |
Collapse
|
22
|
Pentak D, Kozik V, Zieba A, Paździor-Heiske M, Szymczyk A, Jampilek J, Bak A. Preparing a Liposome-Aided Drug Delivery System: The Entrapment and Release Profiles of Doxorubicin and 9-( N-Piperazinyl)-5-methyl-12( H)-quino [3,4-b][1,4]benzothiazinium Chloride with Human Serum Albumin. Pharmaceutics 2025; 17:202. [PMID: 40006569 PMCID: PMC11860059 DOI: 10.3390/pharmaceutics17020202] [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: 12/22/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The principal aim of this work was to prepare a liposomal drug delivery system based on the commercial drug doxorubicin (DOX) and a budding agent with promising anticancer activity, 9-(N-piperazinyl)-5-methyl-12(H)-quino [3,4-b][1,4]benzothiazinium chloride (9-PBThACl). Methods: A spectrophotometric methodology was used to meticulously investigate the drug entrapment and release characteristics of the new liposomal complexes (L) based on dipalmitoylphosphatidylcholine (DPPC) with human serum albumin (HSA) and its defeated analog (dHSA). Results: The impact of the operational parameters (temperature and pH) on the liposome/drug(s)/(d)HSA, namely [LDPPC/9-PBThACl/DOX ]:(d)HSA] systems, as well as the polarity of the phospholipid bilayer, was examined. In order to compare the experimental findings, mathematical models were employed to specify the analytical factors controlling the process of drug release/potential drug release from liposomes. The observed variations in the drug encapsulation and release profiles were due to the combination of liposomal conjugates with human plasma protein. Conclusions: It was proven that changes in the environmental pH directly affect the percentage of drug entrapment in liposomes and the medicine release efficiency. Moreover, the grouping tendency of the liposomal combinations was investigated using a principal component analysis (PCA) and a hierarchical clustering analysis (HCA). Finally, an analysis of variance (ANOVA) confirmed the statistical impact of pH buffering and changing temperature factors on the drug release characteristics of liposomal conjugates.
Collapse
Affiliation(s)
- Danuta Pentak
- Faculty of Chemistry and Pharmacy, University of Opole, Oleska 48, 45-052 Opole, Poland;
| | - Violetta Kozik
- Institute of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland; (V.K.); (M.P.-H.)
| | - Andrzej Zieba
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland;
| | - Marlena Paździor-Heiske
- Institute of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland; (V.K.); (M.P.-H.)
| | - Aleksandra Szymczyk
- Institute of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland; (V.K.); (M.P.-H.)
| | - Josef Jampilek
- Institute of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland; (V.K.); (M.P.-H.)
| | - Andrzej Bak
- Institute of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland; (V.K.); (M.P.-H.)
| |
Collapse
|
23
|
Sancho-Albero M, Decio A, Akpinar R, De Luigi A, Giavazzi R, Terracciano LM, De Cola L. Melanoma extracellular vesicles membrane coated nanoparticles as targeted delivery carriers for tumor and lungs. Mater Today Bio 2025; 30:101433. [PMID: 39866783 PMCID: PMC11764275 DOI: 10.1016/j.mtbio.2024.101433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
Targeting is the most challenging problem to solve for drug delivery systems. Despite the use of targeting units such as antibodies, peptides and proteins to increase their penetration in tumors the amount of therapeutics that reach the target is very small, even with the use of nanoparticles (NPs). Nature has solved the selectivity problem using a combination of proteins and lipids that are exposed on the cell membranes and are able to recognize specific tissues as demonstrated by cancer metastasis. Extracellular vesicles (EVs) have a similar ability in target only certain organs or to return to their original cells, showing home behavior. Here we report a strategy inspired by nature, using a combination of NPs and the targeting cell membranes of EVs. We implement the EV membranes, extracted by the EVs produced by melanoma B16-BL6 cells, as a coating of organosilica porous particles with the aim of targeting tumors and lung metastasis, while avoiding systemic effects and accumulation of the NPs in undesired organs. The tissue-specific fingerprint provided by the EVs-derived membranes from melanoma cells provides preferential uptake into the tumor and selective targeting of lungs. The ability of the EVs hybrid systems to behave as the natural EVs was demonstrated in vitro and in vivo in two different tumor models. As a proof of concept, the loading and release of doxorubicin, was investigated and its accumulation demonstrated in the expected tissues.
Collapse
Affiliation(s)
- María Sancho-Albero
- Department of Biochemistry and Molecular Pharmacology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
| | - Alessandra Decio
- Department of Oncology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
| | - Reha Akpinar
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
| | - Ada De Luigi
- Department of Biochemistry and Molecular Pharmacology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
| | - Raffaella Giavazzi
- Department of Oncology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
| | - Luigi M. Terracciano
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
| | - Luisa De Cola
- Department of Biochemistry and Molecular Pharmacology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
- Department of Pharmaceutical Science, DISFARM. Università degli Studi di Milano, Milan, 20133, Italy
| |
Collapse
|
24
|
Okafor NI, Omoteso OA, Choonara YE. The modification of conventional liposomes for targeted antimicrobial delivery to treat infectious diseases. DISCOVER NANO 2025; 20:19. [PMID: 39883380 PMCID: PMC11782757 DOI: 10.1186/s11671-024-04170-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
Some of the most crucial turning points in the treatment strategies for some major infectious diseases including AIDS, malaria, and TB, have been reached with the introduction of antimicrobials and vaccines. Drug resistance and poor effectiveness are key limitations that need to be overcome. Conventional liposomes have been explored as a delivery system for infectious diseases bioactives to treat infectious diseases to provide an efficient approach to maximize the therapeutic outcomes, drug stability, targetability, to reduce the side-effects of antimicrobials, and enhance vaccine performance where necessary. However, as the pathological understanding of infectious diseases become more known, the need for more advanced liposomal technologies was born to continue having a profound effect on targeted chemotherapy for infectious diseases. This review therefore provides a concise incursion into the most recent and vogue liposomal formulations used to treat infectious diseases. An appraisal of immunological, stimuli-responsive, biomimetic and functionalized liposomes and other novel modifications to conventional liposomes is assimilated in sync with mutations of resistant pathogens.
Collapse
Affiliation(s)
- Nnamdi Ikemefuna Okafor
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | | | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
25
|
Suriyaamporn P, Pornpitchanarong C, Charoenying T, Dechsri K, Ngawhirunpat T, Opanasopit P, Pamornpathomkul B. Artificial intelligence-driven hydrogel microneedle patches integrating 5-fluorouracil inclusion complex-loaded flexible pegylated liposomes for enhanced non-melanoma skin cancer treatment. Int J Pharm 2025; 669:125072. [PMID: 39675535 DOI: 10.1016/j.ijpharm.2024.125072] [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: 07/29/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
The current study focused on the development of crosslinked hydrogel microneedle patches (cHMNs) incorporating 5-FU-hydroxypropyl beta-cyclodextrin inclusion complex-loaded flexible PEGylated liposomes (5-FU-HPβCD-loaded FP-LPs) to enhance treatment efficacy and reduce drug toxicity. The research utilized artificial intelligence (AI) algorithms to design, optimize, and evaluate the cHMNs. Various AI models were assessed for accuracy, with metrics such as root mean square error and coefficient of determination guiding the selection of the most effective formulation. The physicochemical and mechanical properties, swelling behavior, in vitro skin permeation, and safety of the chosen cHMNs were tested. The results demonstrated that the 5-FU-HPβCD-loaded FP-LPs, stabilized with limonene, had an optimal particle size of 36.23 ± 2.42 nm, narrow size distribution, and zeta potential of -10.24 ± 0.37 mV, with high encapsulation efficiency. The cHMNs exhibited a conical needle shape with sufficient mechanical strength to penetrate the stratum corneum up to approximately 467.87 ± 65.12 μm. The system provided a high skin permeation rate of 41.78 ± 4.26 % and significant drug accumulation in the skin. Additionally, the formulation was proven safe in cell culture while effectively inhibiting cancer growth and promoting apoptosis. This study highlights the potential of AI-enhanced cHMNs for delivering 5-FU-HPβCD-loaded FP-LPs transdermally, offering a promising new treatment avenue for non-melanoma skin cancers.
Collapse
Affiliation(s)
- Phuvamin Suriyaamporn
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Chaiyakarn Pornpitchanarong
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Thapakorn Charoenying
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Koranat Dechsri
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Boonnada Pamornpathomkul
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand; Research and Innovation Center for Advanced Therapy Medicinal Products, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
26
|
Yılmaz E, Kacaroglu D, Ozden AK, Aydogan N. Gold nanoparticles decorated FOLFIRINOX loaded liposomes for synergistic therapy of pancreatic cancer. Int J Pharm 2025; 669:125067. [PMID: 39672312 DOI: 10.1016/j.ijpharm.2024.125067] [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: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Pancreatic cancer is predicted to be the second highest cause of cancer deaths by 2030, with a mortality rate of 98 % and a 5-year survival rate of only 4-8 %. FOLFIRINOX which consists of four main ingredients has shown superior efficacy in treating patients with pancreatic cancer compared to other agents and combinations. However, toxicities have prevented full-dose use of FOLFIRINOX. In this study, we present the design of a liposome nanosystem that enables the sequential release of a drug combination that is called FOLFIRINOX using lipid-based nanosystem synergistic chemo/photothermal therapy approaches. The co-eccentric liposome allowed us to locate the drug molecules in different locations giving us the flexibility to release them in a selected order. Core liposome (L2) has a melting temperature of 53.63 °C, it was decorated by gold nanoparticle (L2@AuNP) to bring photothermal responsiveness. The outer liposome structure had a lower melting temperature, which facilitated the sequential release process. The efficacy of photothermal therapy for nanosystem was calculated. The results indicate that coating L2@AuNP nanostructure with L1 liposomes improves efficacy by stabilizing gold nanoparticles. FOLFIRINOX components are encapsulated in a concentric liposome structure according to the order of administration into the body. The concentric liposome structure enables the sequential release of multiple drugs due to the varying phase transition temperatures of the liposomes. The cytotoxic effect of these formulations was evaluated on Panc-1 pancreatic cancer cells; the lowest cell viability was obtained in 4 Liposome(L) under 5 min NIR irradiation. Combination therapy has a higher therapeutic efficacy (70.45 %) when compared to chemotherapy and photothermal therapy used separately. The study's results show the potential of combination therapies to improve therapeutic outcomes, providing a promising path for future research and clinical application.
Collapse
Affiliation(s)
- Emine Yılmaz
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Beytepe, Ankara, Turkey
| | - Demet Kacaroglu
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Ayse Kevser Ozden
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Nihal Aydogan
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Beytepe, Ankara, Turkey; Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara, 06800, Turkey.
| |
Collapse
|
27
|
Pal J, Samanta P, Khan A, Maity R, Mallick AI, Dhara D. Bicontinuous Nanoparticles from Spontaneous Self-Assembly of Block Copolymer Prodrug in Aqueous Medium for Potential Cancer Therapy. ACS Macro Lett 2025; 14:26-34. [PMID: 39693052 DOI: 10.1021/acsmacrolett.4c00590] [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: 12/19/2024]
Abstract
Despite having several advantages, bicontinuously structured polymeric nanoparticles (BSPNPs) are far less explored in the field of controlled drug delivery owing to the requirement of complex precursor copolymers and the associated multistep synthetic procedures. In this work, we report the synthesis of a redox-sensitive diblock copolymer (P1), which was subsequently utilized to prepare doxorubicin (DOX) containing a pH-labile prodrug (P2). P1 and P2 spontaneously self-assembled in aqueous media above their critical aggregation concentration, forming micellar nanoparticles with rare bicontinuous morphology that promotes loading of both hydrophobic and hydrophilic cargoes in different compartments. To the best of our knowledge, the formation of BSPNPs through direct self-assembly in aqueous media has not yet been reported. In vitro cellular studies asserted the higher safety profile of the nanoparticles against noncancerous cells (HEK293T) than free DOX, whereas they displayed higher drug-induced cytotoxicity against cancer cells (MCF-7) in comparison to free DOX, establishing them as promising cancer drug delivery systems.
Collapse
Affiliation(s)
- Juthi Pal
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Pousali Samanta
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Afruja Khan
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741 246, India
| | - Rishabh Maity
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Amirul Islam Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741 246, India
| | - Dibakar Dhara
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| |
Collapse
|
28
|
YALÇIN TE, YETGİN C. Influence of Formulation Composition on the Characteristic Properties of 5-fluorouracil-loaded Liposomes. Turk J Pharm Sci 2025; 21:551-556. [PMID: 39801089 PMCID: PMC11730006 DOI: 10.4274/tjps.galenos.2024.11278] [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: 10/17/2023] [Accepted: 03/04/2024] [Indexed: 01/16/2025]
Abstract
Objectives Variations in the types and quantities of excipients used to prepare liposomes can affect the physicochemical properties of liposome formulations. This study aimed to provide information about the design and fabrication of 5-fluorouracil (5-FU)-loaded liposome formulations using different lipid and cholesterol (CHOL) derivatives. Materials and Methods Passive loading via a small-volume incubation method was used to prepare liposomes. The particle size, polydispersity index, zeta potential, and encapsulation efficiency (EE%) of the formulations were determined. The release studies of the formulations were conducted using a Franz diffusion cell at 37 °C. In this study, a high-pressure liquid chromatography device was used to measure the amount of 5-FU. Results The mean particle sizes of all formulations were between 134 and 166 nm, and they had a negative charge on their surface. Increasing the cholesteryl hemisuccinate content reduced the size of the liposomes. Additionally, all formulations exhibited a low polydispersity index (0.3). The EE% of all formulations exceeded 30%. The in vitro release of 5-FU from liposome formulations followed the Korsemeyer-Peppas model. Conclusion Modifying the lipid and CHOL content in the formulations, as indicated by the experimental results, can change the characteristic properties of liposomes. The use of soybean phosphatidylcholine and cholesteryl hemisuccinate appears to be a promising combination for the preparation of hydrophilic drug-loaded liposome formulations.
Collapse
Affiliation(s)
- Tahir Emre YALÇIN
- Gazi University Faculty of Pharmacy Department of Pharmaceutical Technology, Ankara, Türkiye
| | - Ceren YETGİN
- Gazi University Faculty of Pharmacy Department of Pharmaceutical Technology, Ankara, Türkiye
| |
Collapse
|
29
|
Kodel HDAC, Alizadeh P, Ebrahimi SN, Machado TOX, Oliveira MBPP, Fathi F, Souto EB. Liposomes and Niosomes: New trends and applications in the delivery of bioactive agents for cancer therapy. Int J Pharm 2025; 668:124994. [PMID: 39586512 DOI: 10.1016/j.ijpharm.2024.124994] [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: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Lipid-based nanocarriers have been in continuous development as strategies to enhance drug delivery efficiency. Liposomes are delivery systems primarily composed of phospholipids and cholesterol (or other suitable stabilizers) that have transformed the pharmaceutical field by improving drug targeting and release control. The success of this technology is strongly attributed to phospholipids, which are components of cell membranes, forming a biocompatible system. Nevertheless, drawbacks related to their production cost and stability under certain conditions led to the development of niosomes by replacing phospholipids with non-ionic surfactants. Both liposomes and niosomes have been widely studied and optimized for the delivery of bioactive agents targeting many diseases, including cancer. They can improve the efficacy of cancer therapy by reducing toxicity and off-target effects. Due to the complexity of this disease, many approaches should be considered, and the composition and physical properties of liposomes and niosomes influence the outcomes. In this review, we discuss the role of liposomes and niosomes in delivering bioactives for cancer therapy, emphasizing their specific characteristics, associated challenges, and the latest advancements aimed at enhancing their effectiveness.
Collapse
Affiliation(s)
- Helena de A C Kodel
- Rede Nordeste de Biotecnologia-RENORBIO, University of Tiradentes, Farolândia, 49010-390, Aracaju, Sergipe, Brazil
| | - Paria Alizadeh
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Samad N Ebrahimi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Tatiane O X Machado
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy of University of Porto, Jorge de Viterbo Ferreira, 4050-313, Porto, Portugal; Department of Agroindustry, Federal Institute of Sertão Pernambucano, Campus Petrolina Zona Rural, PE 647, Km 22, PISNC N4, 56302-970, Petrolina, Pernambuco, Brazil
| | - M Beatriz P P Oliveira
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Faezeh Fathi
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Eliana B Souto
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland.
| |
Collapse
|
30
|
Reiten IN, Giraud F, Augedal TT, Førde JL, Moreau P, Gundersen ET, Chapron D, Legrand FX, Anizon F, Herfindal L. Liposomes loaded with daunorubicin and an emetine prodrug for improved selective cytotoxicity towards acute myeloid leukaemia cells. Int J Pharm 2025; 668:124989. [PMID: 39581514 DOI: 10.1016/j.ijpharm.2024.124989] [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: 06/07/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
The backbone of induction therapy in acute myeloid leukaemia (AML) is to use an anthracycline in combination with cytarabine. Despite recent advances in AML therapy, this treatment remains the standard, and it has been largely unchanged for decades. There are few curative options for patients unfit for this treatment. The anti-protozoal agent emetine improves efficacy of anthracycline treatment towards AML in vitro and in vivo but the effect is more potent when emetine is administered 30 minutes after anthracyclines. To delay the onset of protein synthesis inhibition we produced a novel inactive emetine prodrug and co-encapsulated this with the anthracycline daunorubicin (DNR) in liposomes. Nanoencapsulation protects the prodrug from degradation in the blood and ensure simultaneous delivery of both drugs to cancer cells. The prodrug concept will delay the onset of action of emetine relative to DNR. In AML cells, the combination of DNR and the emetine-prodrug in liposomes increased cytotoxicity compared to liposomes with DNR and native emetine. Liposomes loaded with the emetine prodrug did not show increased toxicity towards non-cancerous cell lines and zebrafish larvae. In patients, a liposomal formulation such as that presented herein could allow for a reduced DNR dose without compromising efficacy, thereby reducing toxic side effects and enabling improved therapy for patients not fit for current treatment options.
Collapse
Affiliation(s)
| | - Francis Giraud
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, ICCF, F-63000 Clermont-Ferrand, France
| | - Tuva Torblå Augedal
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Norway
| | - Jan-Lukas Førde
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Pascale Moreau
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, ICCF, F-63000 Clermont-Ferrand, France
| | - Edvin Tang Gundersen
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Norway; Hospital Pharmacies Enterprise, Western Norway, Bergen, Norway
| | - David Chapron
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | | | - Fabrice Anizon
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, ICCF, F-63000 Clermont-Ferrand, France
| | - Lars Herfindal
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Norway.
| |
Collapse
|
31
|
Rahaman W, Chaudhuri A. Self-assembled Lipid Nanoparticles for Killing Triple Negative Breast Cancer Cells. Chem Asian J 2025; 20:e202401049. [PMID: 39466002 DOI: 10.1002/asia.202401049] [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: 08/22/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Triple negative breast cancers (TNBCs) lacking estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) on their cell surfaces are highly aggressive, difficult-to-treat and often relapse. Herein, we report on the self-assembled lipid nanoparticles (LNPs) of two new pegylated lipopeptides for killing TNBCs (MDA-MB-231). The pegylated lipopeptides were synthesized by conjugating an n-hexadecyl hydrophobic tail to one end of a (PEG)27 unit the other distal end of which was covalently grafted with two previously reported tumor targeting RGDK- and CGKRK- peptides. The SEM images of the self-assembled LNPs formed upon dissolution of the pegylated lipopeptides in aqueous medium revealed formation of spherical aggregates. The degree of cellular uptake for the self-assembled LNPs formed by the pegylated CGKRK-lipopeptide were found to be significantly higher than that for the self-assembled LNPs formed by the pegylated RGDK-lipopeptide in MCF-7, MDA-MB-231, HEK-293 and HFF cells. Notably, about 60 % TNBCs (MDA-MB-231 cells) were killed upon treatment with commercially available potent JAK2 inhibitor (WP 1066) loaded LNPs of the pegylated RGDK-lipopeptide. Contrastingly, the same treatment killed only about 20 % non-cancerous HEK-293 cells. The self-assembled pegylated LNPs described herein open the door for undertaking preclinical studies in animal models for TNBCs.
Collapse
Affiliation(s)
- Wahida Rahaman
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Arabinda Chaudhuri
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| |
Collapse
|
32
|
Fatima H, Singh D, Muhammad H, Acharya S, Aziz MA. Improving the use of CRISPR/Cas9 gene editing machinery as a cancer therapeutic tool with the help of nanomedicine. 3 Biotech 2025; 15:17. [PMID: 39711922 PMCID: PMC11656010 DOI: 10.1007/s13205-024-04186-1] [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: 08/02/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-associated protein 9) has revolutionized gene editing tools and paved the way for innovations in medical research for disease diagnosis and treatment. However, better specificity and efficient delivery of this gene machinery make it challenging to successfully edit genes for treating various diseases. This is mainly due to cellular barriers, instability in biological environments, and various off-target effects that prohibit safe and efficient delivery under in vivo conditions. This review examines several delivery modes [plasmid, mRNA, RNP (ribonucleoprotein)] and methods for the CRISPR-Cas9 system delivery, focusing on its potential applications in cancer therapy. Biocompatibility and cytotoxicity are crucial factors determining their safe and effective use. Various nanomaterials have been reviewed for their biocompatibility, limitations, and challenges in treating cancer. Among the reviewed nanoparticles, lipid nanoparticles (LNPs) stand out for their biocompatibility due to their biomimetic lipid bilayer that effectively delivers CRISPR/Cas9 cargoes while reducing toxicity. We discuss challenges in in vivo delivery and associated findings such as encapsulation, target delivery, controlled release, and endosomal escape. Future directions involve addressing limitations and adapting CRISPR-Cas9 for clinical trials, ensuring its safe and effective use.
Collapse
Affiliation(s)
- Hina Fatima
- Polymer and Process Engineering Department, Indian Institute of Technology Roorkee, Uttarakhand, 247001 India
- College of Medicine, Alfaisal University, 11533 Riyadh, Saudi Arabia
| | - Dimple Singh
- Department of Paper Technology, Indian Institute of Technology, Roorkee, Uttarakhand 247001 India
| | - Huzaifa Muhammad
- College of Medicine, Alfaisal University, 11533 Riyadh, Saudi Arabia
| | - Swati Acharya
- Cancer Nanomedicine Lab, Interdisciplinary Nanotechnology Center, Aligarh Muslim University, Aligarh, UP 202002 India
| | - Mohammad Azhar Aziz
- Cancer Nanomedicine Lab, Interdisciplinary Nanotechnology Center, Aligarh Muslim University, Aligarh, UP 202002 India
- Cancer Nanomedicine Consortium, Aligarh Muslim University, Aligarh, UP 202002 India
| |
Collapse
|
33
|
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
|
34
|
Benariba MA, Hannachi K, Zhu S, Zhang Y, Wang X, Zhou N. A liposome-based assay for cancer biomarker detection: exploring the correlation between platelet-derived microvesicles and NSCLC-associated miRNAs. NANOSCALE 2024; 16:22037-22046. [PMID: 39527124 DOI: 10.1039/d4nr03704a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Advances in molecular biology have enabled the identification of numerous cancer biomarkers, offering the potential to improve the diagnosis and prognosis of cancer. In non-small cell lung cancer (NSCLC), the role of platelet-derived microvesicles (PMVs) in cancer progression has received limited attention. While previous studies have focused on the increase of extracellular vesicles in plasma and their interaction with cancer, the expression of microRNAs (miRNAs) delivered through PMVs following platelet activation has remained largely unexplored. This study fills this knowledge gap by investigating miRNA expression in PMVs isolated from healthy donors and NSCLC patients following calcium treatment, a known platelet activator. A significant correlation was found between PMV levels and the expression of specific miRNAs; specifically, miRNA-21 expression increased 7.89 ± 0.44-fold in NSCLC patients and 7.12 ± 0.49-fold in healthy donors after calcium treatment. These findings highlight the potential of PMVs and their miRNA cargo to serve as specific biomarkers for NSCLC, offering valuable insights into cancer diagnosis and prognosis. To facilitate the sensitive detection of these miRNAs, a novel carboxyfluorescein (CF)-loaded liposome-based assay was developed. This assay demonstrated enhanced sensitivity, achieving a detection limit of 1.03 pg mL-1, when combined with a calcium platelet-activation approach. This research has the potential to lead to the development of innovative diagnostic tools and therapeutic strategies, ultimately improving outcomes for patients with NSCLC and other cancers.
Collapse
Affiliation(s)
- Mohamed Aimene Benariba
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
- Bioengineering Laboratory, Ecole Nationale Supérieure de Biotechnologie, Ville Universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria
| | - Kanza Hannachi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Sha Zhu
- Department of Urology, the Wuxi No. 2 People's Hospital, Jiangnan University Medical Center, Wuxi 214002, China.
| | - Yuting Zhang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Xiaoli Wang
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| | - Nandi Zhou
- School of Biotechnology and Key Laboratory of Carbohydrate Chemistry and Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
35
|
Koochaki R, Amini E, Zarehossini S, Zareh D, Haftcheshmeh SM, Jha SK, Kesharwani P, Shakeri A, Sahebkar A. Alkaloids in Cancer therapy: Targeting the tumor microenvironment and metastasis signaling pathways. Fitoterapia 2024; 179:106222. [PMID: 39343104 DOI: 10.1016/j.fitote.2024.106222] [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: 02/07/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
The use of phytomedicine in cancer therapy is a growing field of research that takes use of the medicinal properties of plant-derived compounds. Under the domain of cancer therapy and management, alkaloids, a prominent group of natural compounds, have showed significant potential. Alkaloids often affect a wide range of essential cellular mechanisms involved in cancer progression. These multi-targeting capabilities, can give significant advantages to alkaloids in overcoming resistance mechanisms. For example, berberine, an alkaloid found in Berberis species, is widely reported to induce apoptosis by activating caspases and regulating apoptotic pathways. Notably, alkaloids like as quinine have showed promise in inhibiting the formation of new blood vessels required for tumor growth. In addition, alkaloids have shown anti-proliferative and anticancer properties mostly via modulating key signaling pathways involved in metastasis, including those regulating epithelial-mesenchymal transition. This work provides a comprehensive overview of naturally occurring alkaloids that exhibit anticancer properties, with a specific emphasis on their underlying molecular mechanisms of action. Furthermore, many methods to modify previously reported difficult physicochemical properties using nanocarriers in order to enhance its systemic bioavailability have been discussed as well. This study also includes information on newly discovered alkaloids that are now being studied in clinical trials for their potential use in cancer treatment. Further, we have also briefly mentioned on the application of high-throughput screening and molecular dynamics simulation for acceleration on the identification of potent alkaloids based compounds to target and treat cancer.
Collapse
Affiliation(s)
- Raoufeh Koochaki
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Elaheh Amini
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Sara Zarehossini
- Department of Cell & Molecular Biology (genetic), Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Danial Zareh
- Department of Cell & Molecular Biology (genetic), Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Abolfazl Shakeri
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran..
| |
Collapse
|
36
|
Dejeu IL, Vicaș LG, Marian E, Ganea M, Frenț OD, Maghiar PB, Bodea FI, Dejeu GE. Innovative Approaches to Enhancing the Biomedical Properties of Liposomes. Pharmaceutics 2024; 16:1525. [PMID: 39771504 PMCID: PMC11728823 DOI: 10.3390/pharmaceutics16121525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Liposomes represent a promising class of drug delivery systems that enhance the therapeutic efficacy and safety of various pharmaceutical agents. Also, they offer numerous advantages compared to traditional drug delivery methods, including targeted delivery to specific sites, controlled release, and fewer side effects. This review meticulously examines the methodologies employed in the preparation and characterization of liposomal formulations. With the rising incidence of adverse drug reactions, there is a pressing need for innovative delivery strategies that prioritize selectivity, specificity, and safety. Nanomedicine promises to revolutionize diagnostics and treatments, addressing current limitations and improving disease management, including cancer, which remains a major global health challenge. This paper aims to conduct a comprehensive study on the interest of biomedical research regarding nanotechnology and its implications for further applications.
Collapse
Affiliation(s)
- Ioana Lavinia Dejeu
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Laura Grațiela Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Mariana Ganea
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Olimpia Daniela Frenț
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Paula Bianca Maghiar
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania; (P.B.M.); (F.I.B.)
| | - Flaviu Ionut Bodea
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania; (P.B.M.); (F.I.B.)
| | - George Emanuiel Dejeu
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 Piata 1 Decembrie Street, 410073 Oradea, Romania;
| |
Collapse
|
37
|
Shakori Poshteh S, Alipour S, Varamini P. Harnessing curcumin and nanotechnology for enhanced treatment of breast cancer bone metastasis. DISCOVER NANO 2024; 19:177. [PMID: 39527354 PMCID: PMC11554965 DOI: 10.1186/s11671-024-04126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Breast cancer (BC) bone metastasis poses a significant clinical challenge due to its impact on patient prognosis and quality of life. Curcumin (CUR), a natural polyphenol compound found in turmeric, has shown potential in cancer therapy due to its anti-inflammatory, antioxidant, and anticancer properties. However, its metabolic instability and hydrophobicity have hindered its clinical applications, leading to a short plasma half-life, poor absorption, and low bioavailability. To enhance the drug-like properties of CUR, nanotechnology-based delivery strategies have been employed, utilizing polymeric, lipidic, and inorganic nanoparticles (NPs). These approaches have effectively overcome CUR's inherent limitations by enhancing its stability and cellular bioavailability both in vitro and in vivo. Moreover, targeting molecules with high selectivity towards bone metastasized breast cancer cells can be used for site specific delivery of curcumin. Alendronate (ALN), a bone-seeking bisphosphonate, is one such moiety with high selectivity towards bone and thus can be effectively used for targeted delivery of curcumin loaded nanocarriers. This review will detail the process of bone metastasis in BC, elucidate the mechanism of action of CUR, and assess the efficacy of nanotechnology-based strategies for CUR delivery. Specifically, it will focus on how these strategies enhance CUR's stability and improve targeted delivery approaches in the treatment of BC bone metastasis.
Collapse
Affiliation(s)
- Shiva Shakori Poshteh
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Shohreh Alipour
- Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Drug and Food Control, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Pegah Varamini
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
- The University of Sydney Nano Institute, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
38
|
Chatterjee D, Bhattacharya S, Kumari L, Datta A. Aptamers: ushering in new hopes in targeted glioblastoma therapy. J Drug Target 2024; 32:1005-1028. [PMID: 38923419 DOI: 10.1080/1061186x.2024.2373306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Glioblastoma, a formidable brain cancer, has remained a therapeutic challenge due to its aggressive nature and resistance to conventional treatments. Recent data indicate that aptamers, short synthetic DNA or RNA molecules can be used in anti-cancer therapy due to their better tumour penetration, specific binding affinity, longer retention in tumour sites and their ability to cross the blood-brain barrier. With the ability to modify these oligonucleotides through the selection process, and using rational design to modify them, post-SELEX aptamers offer several advantages in glioblastoma treatment, including precise targeting of cancer cells while sparing healthy tissue. This review discusses the pivotal role of aptamers in glioblastoma therapy and diagnosis, emphasising their potential to enhance treatment efficacy and also highlights recent advancements in aptamer-based therapies which can transform the landscape of glioblastoma treatment, offering renewed hope to patients and clinicians alike.
Collapse
Affiliation(s)
- Debarpan Chatterjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Srijan Bhattacharya
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Leena Kumari
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Aparna Datta
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| |
Collapse
|
39
|
Sorrentino C, Ciummo SL, Fieni C, Di Carlo E. Nanomedicine for cancer patient-centered care. MedComm (Beijing) 2024; 5:e767. [PMID: 39434967 PMCID: PMC11491554 DOI: 10.1002/mco2.767] [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: 07/12/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Cancer is a leading cause of morbidity and mortality worldwide, and an increase in incidence is estimated in the next future, due to population aging, which requires the development of highly tolerable and low-toxicity cancer treatment strategies. The use of nanotechnology to tailor treatments according to the genetic and immunophenotypic characteristics of a patient's tumor, and to allow its targeted release, can meet this need, improving the efficacy of treatment and minimizing side effects. Nanomedicine-based approach for the diagnosis and treatment of cancer is a rapidly evolving field. Several nanoformulations are currently in clinical trials, and some have been approved and marketed. However, their large-scale production and use are still hindered by an in-depth debate involving ethics, intellectual property, safety and health concerns, technical issues, and costs. Here, we survey the key approaches, with specific reference to organ-on chip technology, and cutting-edge tools, such as CRISPR/Cas9 genome editing, through which nanosystems can meet the needs for personalized diagnostics and therapy in cancer patients. An update is provided on the nanopharmaceuticals approved and marketed for cancer therapy and those currently undergoing clinical trials. Finally, we discuss the emerging avenues in the field and the challenges to be overcome for the transfer of nano-based precision oncology into clinical daily life.
Collapse
Affiliation(s)
- Carlo Sorrentino
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Stefania Livia Ciummo
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Cristiano Fieni
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| |
Collapse
|
40
|
Shahzad A, Teng Z, Yameen M, Liu W, Cui K, Liu X, Sun Y, Duan Q, Xia J, Dong Y, Bai Z, Peng D, Zhang J, Xu Z, Pi J, Yang Z, Zhang Q. Innovative lipid nanoparticles: A cutting-edge approach for potential renal cell carcinoma therapeutics. Biomed Pharmacother 2024; 180:117465. [PMID: 39321512 DOI: 10.1016/j.biopha.2024.117465] [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: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024] Open
Abstract
The kidney plays a crucial role in regulating homeostasis within the human body. Renal cell carcinoma (RCC) is the most common form of kidney cancer, accounting for nearly 90 % of all renal malignancies. Despite the availability of various therapeutic strategies, RCC remains a challenging disease due to its resistance to conventional treatments. Nanotechnology has emerged as a promising field, offering new opportunities in cancer therapeutics. It presents several advantages over traditional methods, enabling diverse biomedical applications, including drug delivery, prevention, diagnosis, and treatment. Lipid nanoparticles (LNPs), approximately 100 nm in size, are derived from a range of lipids and other biochemical compounds. these particulates are designed to overcome biological barriers, allowing them to selectively accumulate at diseased target sites for effective therapeutic action. Many pharmaceutically important compounds face challenges such as poor solubility in aqueous solutions, chemical and physiological instability, or toxicity. LNP technology stands out as a promising drug delivery system for bioactive organic compounds. This article reviews the applications of LNPs in RCC treatment and explores their potential clinical translation, identifying the most viable LNPs for medical use. With ongoing advancement in LNP-based anticancer strategies, there is a growing potential to improve the management and treatment of renal cancer.
Collapse
Affiliation(s)
- Asif Shahzad
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Zhuoran Teng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Muhammad Yameen
- Department of Biochemistry, Government College University Faisalabad, Punjab 38000, Pakistan
| | - Wenjing Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Kun Cui
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Xiangjie Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yijian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Qiuxin Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - JiaoJiao Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yurong Dong
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Ziyuan Bai
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Dongmei Peng
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jinshan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Zhe Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Zhe Yang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| | - Qiao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, China.
| |
Collapse
|
41
|
Mod Razif MRF, Chan SY, Chew YL, Hassan M, Ahmad Hisham S, Abdul Rahman S, Mai CW, Teo MYM, Kee PE, Khoo KS, Lee SK, Liew KB. Recent Developments in Luteolin-Loaded Nanoformulations for Enhanced Anti-Carcinogenic Activities: Insights from In Vitro and In Vivo Studies. SCI 2024; 6:68. [DOI: 10.3390/sci6040068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2025] Open
Abstract
With approximately 18 million people affected by cancer in 2020 globally, scientists are exploring innovative approaches to develop effective treatments for various types of cancer. Traditional chemotherapy drugs, although effective against cancer cells, often lead to significant side effects on healthy tissues, such as hair loss, anemia, and nausea. To discover safer alternatives, researchers are investigating natural bioactive compounds found abundantly in plants. Luteolin, a flavonoid found in celery and artichokes, stands out due to its diverse anti-carcinogenic properties, including inhibiting proliferation, inducing apoptosis, activating autophagy, and inhibiting angiogenesis and metastasis. However, the therapeutic potential of luteolin is hindered by challenges related to its bioavailability and solubility. This critical review explores the specific anti-carcinogenic properties of luteolin while analyzing the impact of its limited bioavailability and solubility on effectiveness. Additionally, it investigates the outcomes of encapsulating luteolin in nanoformulations, providing insights into potential strategies for enhancing its anti-carcinogenic effects. Finally, the review compares the efficacy of luteolin with and without nanoformulations. This review provides valuable insights into the potential of utilizing luteolin-loaded nanoformulations as a safer and more effective method for treating cancer, contributing to the ongoing efforts in improving cancer care and outcomes worldwide.
Collapse
Affiliation(s)
| | - Siok Yee Chan
- School of Pharmaceutical Science, Universiti Sains Malaysia, Jalan Universiti, Gelugor 11700, PNG, Malaysia
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, UCSI Heights, Jalan Puncak Menara Gading, Taman Connaught, Cheras 56000, KUL, Malaysia
| | - Masriana Hassan
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Jalan Universiti 1, Serdang 43400, SGR, Malaysia
| | - Shairyzah Ahmad Hisham
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya 63000, SGR, Malaysia
| | - Shamima Abdul Rahman
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya 63000, SGR, Malaysia
| | - Chun-Wai Mai
- Faculty of Pharmaceutical Sciences, UCSI University, UCSI Heights, Jalan Puncak Menara Gading, Taman Connaught, Cheras 56000, KUL, Malaysia
| | - Michelle Yee Mun Teo
- Faculty of Applied Sciences, UCSI University, UCSI Heights, Jalan Puncak Menara Gading, Taman Connaught, Cheras 56000, KUL, Malaysia
| | - Phei Er Kee
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chungli, Taoyuan 320, Taiwan
| | - Kuan Shiong Khoo
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chungli, Taoyuan 320, Taiwan
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, TN, India
| | - Siew-Keah Lee
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Kajang 43000, SGR, Malaysia
| | - Kai Bin Liew
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya 63000, SGR, Malaysia
| |
Collapse
|
42
|
Pont I, Felipe R, Frías JC, Chicote JU, García-España A, García-España E, Albelda MT. An Effective Liposome-Based Nanodelivery System for Naphthalene Derivative Polyamines with Antitumor Activity. Biomolecules 2024; 14:1347. [PMID: 39595524 PMCID: PMC11591986 DOI: 10.3390/biom14111347] [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: 09/24/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024] Open
Abstract
This study focuses on the development of a novel liposome-based nanodelivery system designed to encapsulate polyamine-1, a compound with potential anti-tumor properties. The main objective of this work was to enhance the therapeutic and imaging potential of polyamine-1 by incorporating it into liposome-based nanoparticles, which were functionalized with a gadolinium complex for imaging purposes and a fluorescent phospholipid for tracking applications. These nanoparticles were characterized by measuring their size, shape, polydispersity index, zeta potential and encapsulation efficiency. In vitro experiments were conducted to evaluate the antitumor activity, specifically determining the cytotoxicity of both free and encapsulated polyamine-1 in cancerous and non-cancerous cell lines. Additionally, the study shows the enhanced signal intensity of gadolinium-loaded liposomes by T1-weighted MRI, highlighting their imaging potential. The experimental results suggest that this liposome-based nanodelivery system not only has therapeutic potential in targeted cancer therapy but also could be advantageous for diagnostic imaging, particularly in MRI applications.
Collapse
Affiliation(s)
- Isabel Pont
- Instituto de Ciencia Molecular, Departamento de Química Inorgánica, Universidad de Valencia, 46010 Valencia, Spain; (I.P.); (R.F.); (E.G.-E.)
| | - Rubén Felipe
- Instituto de Ciencia Molecular, Departamento de Química Inorgánica, Universidad de Valencia, 46010 Valencia, Spain; (I.P.); (R.F.); (E.G.-E.)
| | - Juan C. Frías
- Departamento de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, Spain;
| | - Javier U. Chicote
- Unitat de Recerca, Hospital Joan XXIII, Institut de Investigació Sanitaria Pere Virgili (IISPV), Universitat Roviri i Virgili, 43002 Tarragona, Spain;
| | - Antonio García-España
- Unitat de Recerca, Hospital Joan XXIII, Institut de Investigació Sanitaria Pere Virgili (IISPV), Universitat Roviri i Virgili, 43002 Tarragona, Spain;
| | - Enrique García-España
- Instituto de Ciencia Molecular, Departamento de Química Inorgánica, Universidad de Valencia, 46010 Valencia, Spain; (I.P.); (R.F.); (E.G.-E.)
| | - M. Teresa Albelda
- Department of Inorganic Chemistry, University of Valencia, 46010 Burjassot, Spain
| |
Collapse
|
43
|
Alexandru I, Davidescu L, Motofelea AC, Ciocarlie T, Motofelea N, Costachescu D, Marc MS, Suppini N, Șovrea AS, Coșeriu RL, Bondor DA, Bobeică LG, Crintea A. Emerging Nanomedicine Approaches in Targeted Lung Cancer Treatment. Int J Mol Sci 2024; 25:11235. [PMID: 39457017 PMCID: PMC11508987 DOI: 10.3390/ijms252011235] [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: 07/30/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Lung cancer, the leading cause of cancer-related deaths worldwide, is characterized by its aggressive nature and poor prognosis. As traditional chemotherapy has the disadvantage of non-specificity, nanomedicine offers innovative approaches for targeted therapy, particularly through the development of nanoparticles that can deliver therapeutic agents directly to cancer cells, minimizing systemic toxicity and enhancing treatment efficacy. VEGF and VEGFR are shown to be responsible for activating different signaling cascades, which will ultimately enhance tumor development, angiogenesis, and metastasis. By inhibiting VEGF and VEGFR signaling pathways, these nanotherapeutics can effectively disrupt tumor angiogenesis and proliferation. This review highlights recent advancements in nanoparticle design, including lipid-based, polymeric, and inorganic nanoparticles, and their clinical implications in improving lung cancer outcomes, exploring the role of nanomedicine in lung cancer diagnoses and treatment.
Collapse
Affiliation(s)
- Isaic Alexandru
- Department X of General Surgery, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Lavinia Davidescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Alexandru Cătălin Motofelea
- Department of Internal Medicine, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Tudor Ciocarlie
- Department VII Internal Medicine II, Discipline of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Nadica Motofelea
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania;
| | - Dan Costachescu
- Radiology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Monica Steluta Marc
- Discipline of Pulmonology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (M.S.M.); (N.S.)
| | - Noemi Suppini
- Discipline of Pulmonology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (M.S.M.); (N.S.)
| | - Alina Simona Șovrea
- Department of Morphological Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Răzvan-Lucian Coșeriu
- Department of Microbiology, University of Medicine, Pharmacy, Science and Technology “George Emil Palade”, 540142 Târgu-Mures, Romania;
| | - Daniela-Andreea Bondor
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| | - Laura-Gabriela Bobeică
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| | - Andreea Crintea
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| |
Collapse
|
44
|
Castillo Cruz B, Chinapen Barletta S, Ortiz Muñoz BG, Benitez-Reyes AS, Amalbert Perez OA, Cardona Amador AC, Vivas-Mejia PE, Barletta GL. Effect of Cyclodextrins Formulated in Liposomes and Gold and Selenium Nanoparticles on siRNA Stability in Cell Culture Medium. Pharmaceuticals (Basel) 2024; 17:1344. [PMID: 39458985 PMCID: PMC11510567 DOI: 10.3390/ph17101344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Encapsulation of siRNA fragments inside liposome vesicles has emerged as an effective method for delivering siRNAs in vitro and in vivo. However, the liposome's fluid-phospholipid bilayer of liposomes allows siRNA fragments to diffuse out of the liposome, decreasing the dose concentration and therefore the effectiveness of the carrier. We have previously reported that β-cyclodextrins formulated in liposomes help increase the stability of siRNAs in cell culture medium. Here, we continued that study to include α, γ, methyl-β-cyclodextrins and β-cyclodextrin-modified gold and selenium nanoparticles. METHODS We used Isothermal Titration Calorimetry to study the binding thermodynamics of siRNAs to the cyclodextrin-modified nanoparticles and to screen for the best adamantane derivative to modify the siRNA fragments, and we used gel electrophoresis to study the stabilization effect of siRNA by cyclodextrins and the nanoparticles. RESULTS We found that only β- and methyl-β-cyclodextrins increased siRNA serum stability. Cyclodextrin-modified selenium nanoparticles also stabilize siRNA fragments in serum, and siRNAs chemically modified with an adamantane moiety (which forms inclusion complexes with the cyclodextrin-modified-nanoparticles) show a strong stabilization effect. CONCLUSIONS β-cyclodextrins are good additives to stabilize siRNA in cell culture medium, and the thermodynamic data we generated of the interaction between cyclodextrins and adamantane analogs (widely used in drug delivery studies), should serve as a guide for future studies where cyclodextrins are sought for the delivery and solvation of small organic molecules.
Collapse
Affiliation(s)
- Betzaida Castillo Cruz
- Department of Chemistry, University of Puerto Rico at Humacao, Humacao 00791, Puerto Rico; (B.C.C.); (B.G.O.M.); (A.S.B.-R.); (O.A.A.P.); (A.C.C.A.)
| | - Sandra Chinapen Barletta
- Department of Physiology/Pathology, San Juan Bautista School of Medicine, Caguas 00725, Puerto Rico;
| | - Bryan G. Ortiz Muñoz
- Department of Chemistry, University of Puerto Rico at Humacao, Humacao 00791, Puerto Rico; (B.C.C.); (B.G.O.M.); (A.S.B.-R.); (O.A.A.P.); (A.C.C.A.)
| | - Adriana S. Benitez-Reyes
- Department of Chemistry, University of Puerto Rico at Humacao, Humacao 00791, Puerto Rico; (B.C.C.); (B.G.O.M.); (A.S.B.-R.); (O.A.A.P.); (A.C.C.A.)
| | - Omar A. Amalbert Perez
- Department of Chemistry, University of Puerto Rico at Humacao, Humacao 00791, Puerto Rico; (B.C.C.); (B.G.O.M.); (A.S.B.-R.); (O.A.A.P.); (A.C.C.A.)
| | - Alexander C. Cardona Amador
- Department of Chemistry, University of Puerto Rico at Humacao, Humacao 00791, Puerto Rico; (B.C.C.); (B.G.O.M.); (A.S.B.-R.); (O.A.A.P.); (A.C.C.A.)
| | - Pablo E. Vivas-Mejia
- Department of Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan 0035, Puerto Rico
- Comprehensive Cancer Center, University of Puerto Rico, San Juan 00936, Puerto Rico
| | - Gabriel L. Barletta
- Department of Chemistry, University of Puerto Rico at Humacao, Humacao 00791, Puerto Rico; (B.C.C.); (B.G.O.M.); (A.S.B.-R.); (O.A.A.P.); (A.C.C.A.)
| |
Collapse
|
45
|
Hazarika D, Sarma S, Shankarishan P. Nanotechnology in cancer therapeutics, diagnosis, and management. BIOTECHNOLOGIA 2024; 105:287-303. [PMID: 39439717 PMCID: PMC11492894 DOI: 10.5114/bta.2024.141807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 10/25/2024] Open
Abstract
Nanotechnology presents an exciting opportunity in cancer research by offering significant advancements in therapies, diagnosis, and management. It possesses unparalleled potential to enhance the accuracy and effectiveness of cancer therapy while simultaneously reducing adverse effects, owing to its distinctive capability to manipulate matter at a molecular level. Using nanoparticle carriers has facilitated the precise administration of therapeutic agents to afflicted areas within the human body through customized drug delivery systems, resulting in improved treatment accuracy and efficacy while reducing adverse effects. These techniques improve drug solubility and stability, leading to elevated levels of biochemical availability and improved efficacy outcomes for patients with minimal negative effects during treatment cycles. Another use case for nanoparticles includes tumor imaging; functionalized with targeting ligands containing diagnostic agents, they foster early detection, making quicker remedial action plans possible. Overall, the incorporation of nanotechnology ensures a promising future, although it stresses the need to address regulatory hurdles and safety concerns before widespread clinical implementation. Despite the complexity of cancer research and patient care, nanotechnology shows promise in transforming both fields.
Collapse
Affiliation(s)
- Disha Hazarika
- University of Science and Technology Meghalaya (USTM), Meghalaya, India
| | - Sumit Sarma
- University of Science and Technology Meghalaya (USTM), Meghalaya, India
| | | |
Collapse
|
46
|
Chaudhary AA, Fareed M, Khan SUD, Alneghery LM, Aslam M, Alex A, Rizwanullah M. Exploring the therapeutic potential of lipid-based nanoparticles in the management of oral squamous cell carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1223-1246. [PMID: 39465011 PMCID: PMC11502080 DOI: 10.37349/etat.2024.00272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a highly malignant and invasive tumor with significant mortality and morbidity. Current treatment modalities such as surgery, radiotherapy, and chemotherapy encounter significant limitations, such as poor targeting, systemic toxicity, and drug resistance. There is an urgent need for novel therapeutic strategies that offer targeted delivery, enhanced efficacy, and reduced side effects. The advent of lipid-based nanoparticles (LNPs) offers a promising tool for OSCC therapy, potentially overcoming the limitations of current therapeutic approaches. LNPs are composed of biodegradable and biocompatible lipids, which minimize the risk of toxicity and adverse effects. LNPs can encapsulate hydrophobic drugs, improving their solubility and stability in the biological environment, thereby enhancing their bioavailability. LNPs demonstrate significantly higher ability to encapsulate lipophilic drugs than other nanoparticle types. LNPs offer excellent storage stability, minimal drug leakage, and controlled drug release, making them highly effective nanoplatforms for the delivery of chemotherapeutic agents. Additionally, LNPs can be modified by complexing them with specific target ligands on their surface. This surface modification allows the active targeting of LNPs to the tumors in addition to the passive targeting mechanism. Furthermore, the PEGylation of LNPs improves their hydrophilicity and enhances their biological half-life by reducing clearance by the reticuloendothelial system. This review aims to discuss current treatment approaches and their limitations, as well as recent advancements in LNPs for better management of OSCC.
Collapse
Affiliation(s)
- Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammad Fareed
- College of Medicine, AlMaarefa University, Diriyah, Riyadh 11597, Saudi Arabia
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Lina M Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001, Kurdistan Region, Iraq
| | - Arockia Alex
- Molecular and Nanobiotechnology Laboratory (MNBL), Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Md Rizwanullah
- Drug Delivery and Nanomedicine Unit, Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| |
Collapse
|
47
|
Duan M, Cao R, Yang Y, Chen X, Liu L, Ren B, Wang L, Goh BC. Blood-Brain Barrier Conquest in Glioblastoma Nanomedicine: Strategies, Clinical Advances, and Emerging Challenges. Cancers (Basel) 2024; 16:3300. [PMID: 39409919 PMCID: PMC11475686 DOI: 10.3390/cancers16193300] [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: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is a prevalent type of malignancy within the central nervous system (CNS) that is associated with a poor prognosis. The standard treatment for GBM includes the surgical resection of the tumor, followed by radiotherapy and chemotherapy; yet, despite these interventions, overall treatment outcomes remain suboptimal. The blood-brain barrier (BBB), which plays a crucial role in maintaining the stability of brain tissue under normal physiological conditions of the CNS, also poses a significant obstacle to the effective delivery of therapeutic agents to GBMs. Recent preclinical studies have demonstrated that nanomedicine delivery systems (NDDSs) offer promising results, demonstrating both effective GBM targeting and safety, thereby presenting a potential solution for targeted drug delivery. In this review, we first explore the various strategies employed in preclinical studies to overcome the BBB for drug delivery. Subsequently, the results of the clinical translation of NDDSs are summarized, highlighting the progress made. Finally, we discuss potential strategies for advancing the development of NDDSs and accelerating their translational research through well-designed clinical trials in GBM therapy.
Collapse
Affiliation(s)
- Mengyun Duan
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Ruina Cao
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China;
| | - Yuan Yang
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Xiaoguang Chen
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lian Liu
- Department of Pharmacology, Health Science Center, Yangtze University, Jingzhou 434023, China;
| | - Boxu Ren
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Boon-Cher Goh
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
48
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
49
|
Corte-Real M, Veiga F, Paiva-Santos AC, Pires PC. Improving Skin Cancer Treatment by Dual Drug Co-Encapsulation into Liposomal Systems-An Integrated Approach towards Anticancer Synergism and Targeted Delivery. Pharmaceutics 2024; 16:1200. [PMID: 39339235 PMCID: PMC11434718 DOI: 10.3390/pharmaceutics16091200] [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: 08/13/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Skin cancer is a high-incidence complex disease, representing a significant challenge to public health, with conventional treatments often having limited efficacy and severe side effects. Nanocarrier-based systems provide a controlled, targeted, and efficacious methodology for the delivery of therapeutic molecules, leading to enhanced therapeutic efficacy, the protection of active molecules from degradation, and reduced adverse effects. These features are even more relevant in dual-loaded nanosystems, with the encapsulated drug molecules leading to synergistic antitumor effects. This review examines the potential of improving the treatment of skin cancer through dual-loaded liposomal systems. The performed analysis focused on the characterization of the developed liposomal formulations' particle size, polydispersity index, zeta potential, encapsulation efficiency, drug release, and in vitro and/or in vivo therapeutic efficacy and safety. The combination of therapeutic agents such as doxorubicin, 5-fluorouracil, paclitaxel, cetuximab, celecoxib, curcumin, resveratrol, quercetin, bufalin, hispolon, ceramide, DNA, STAT3 siRNA, Bcl-xl siRNA, Aurora-A inhibitor XY-4, 1-Methyl-tryptophan, and cytosine-phosphate-guanosine anionic peptide led to increased and targeted anticancer effects, having relevant complementary effects as well, including antioxidant, anti-inflammatory, and immunomodulatory activities, all relevant in skin cancer pathophysiology. The substantial potential of co-loaded liposomal systems as highly promising for advancing skin cancer treatment is demonstrated.
Collapse
Affiliation(s)
- Margarida Corte-Real
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal (F.V.)
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal (F.V.)
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal (F.V.)
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Patrícia C. Pires
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal (F.V.)
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
50
|
Sharma S, Chakraborty M, Yadav D, Dhullap A, Singh R, Verma RK, Bhattacharya S, Singh S. Strategic Developments in Polymer-Functionalized Liposomes for Targeted Colon Cancer Therapy: An Updated Review of Clinical Trial Data and Future Horizons. Biomacromolecules 2024; 25:5650-5669. [PMID: 39162323 DOI: 10.1021/acs.biomac.4c00847] [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: 08/21/2024]
Abstract
Liposomes, made up of phospholipid bilayers, are efficient nanocarriers for drug delivery because they can encapsulate both hydrophilic and lipophilic drugs. Conventional cancer treatments sometimes involve considerable toxicities and adverse drug reactions (ADRs), which limits their clinical value. Despite liposomes' promise in addressing these concerns, clinical trials have revealed significant limitations, including stability, targeted distribution, and scaling challenges. Recent clinical trials have focused on enhancing liposome formulations to increase therapeutic efficacy while minimizing negative effects. Notably, the approval of liposomal medications like Doxil demonstrates their potential in cancer treatment. However, the intricacy of liposome preparation and the requirement for comprehensive regulatory approval remain substantial impediments. Current clinical trial updates show continued efforts to improve liposome stability, targeting mechanisms, and payload capacity in order to address these issues. The future of liposomal drug delivery in cancer therapy depends on addressing these challenges in order to provide patients with more effective and safer treatment alternatives.
Collapse
Affiliation(s)
- Satyam Sharma
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| | - Moitrai Chakraborty
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| | - Dharmendra Yadav
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| | - Aniket Dhullap
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| | - Raghuraj Singh
- Pharmaceutical Nanotechnology lab, Institute of Nano Science and Technology (INST), Sector 81, Mohali, Punjab 140306, India
| | - Rahul Kumar Verma
- Pharmaceutical Nanotechnology lab, Institute of Nano Science and Technology (INST), Sector 81, Mohali, Punjab 140306, India
| | - Sankha Bhattacharya
- SVKM's NMIMS School of Pharmacy & Technology Management, Shirpur, Dist. Dhule, Maharashtra 425405, India
| | - Sanjiv Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| |
Collapse
|