1
|
Ali MS, Gupta G, Alsayari A, Wahab S, Kesharwani P. Biotinylated nanoparticles: A new frontier in nanomedicine and targeted cancer therapy. BIOMATERIALS ADVANCES 2025; 176:214366. [PMID: 40479769 DOI: 10.1016/j.bioadv.2025.214366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 05/11/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025]
Abstract
The development of targeted drug delivery systems has become a cornerstone of modern cancer therapy which provides a pathway to maximize treatment effectiveness while reducing side effects. Among the plethora of innovative strategies, biotinylated nanoparticles have evolved as a hopeful tool due to their ability to exploit the elevated expression of biotin receptors on cancerous cells. The design, synthesis, and functionalization of biotinylated nanoparticles for cancer treatment are thoroughly examined in this review article. By leveraging biotin's high affinity for biotin receptors, these nanoparticles achieve selective cancerous cell targeting, leading to enhanced drug bioavailability and cellular uptake. The discussion extends to the underlying mechanisms of drug release, receptor-mediated endocytosis, and strategies for achieving endosomal escape or pH-sensitive drug activation. Furthermore, the article also emphasizes how biotinylation in combination therapy allows for synergistic effects with immunomodulators, nucleic acids, and chemotherapeutic drugs. Preclinical studies are examined to underscore the translational potential of these systems. The review concludes by addressing current challenges, including scalability, and potential immunogenicity, while proposing future directions for optimizing biotinylated nanoparticles as a transformative approach in cancer treatment.
Collapse
Affiliation(s)
- Mohd Shoab Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Garima Gupta
- Graphic Era Hill University, Dehradun 248002, India; School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| |
Collapse
|
2
|
Ghosh R, Kumar M, Kumar S, Komal K, Sharma R, Kurmi BD. Small molecule therapeutics for receptor-mediated targeting through liposomes in breast cancer treatment: A comprehensive review. Bioorg Chem 2025; 160:108442. [PMID: 40199009 DOI: 10.1016/j.bioorg.2025.108442] [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: 01/24/2025] [Revised: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Breast cancer (BC) remains a significant global health challenge, with conventional treatment approaches such as surgery, chemotherapy, and radiation therapy. These approaches face limitations in targeting, toxicity, and efficacy. Liposomal drug delivery systems have emerged as promising tools for targeted breast cancer therapies. Liposomes can encapsulate both hydrophilic and hydrophobic drugs, improve drug distribution, and reduce the side effects. Passive targeting exploits the enhanced permeability and retention effect in tumor tissues, whereas active targeting employs small molecule ligands such as aptamers, folic acid (FA), transferrin, and monoclonal antibodies to specifically bind to overexpressed receptors on cancer cells. Aptamer-functionalized liposomes exhibit high specificity and affinity, folate and transferrin receptor targeting enhances cellular uptake and cytotoxicity, and antibody-conjugated liposomes improve drug delivery and efficacy by targeting specific antigens. Dual-responsive liposomes are sensitive to multiple stimuli and further enhance targeting precision. However, challenges remain, including tumor heterogeneity, limited penetration, and potential immunogenicity. Current research has focused on developing stable and effective formulations and exploring combination-targeting strategies to overcome these limitations. With further advancements, targeted liposomal drug delivery systems hold great promise in improving breast cancer treatment outcomes and reducing adverse effects.
Collapse
Affiliation(s)
- Rashmi Ghosh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India.
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Rohit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| |
Collapse
|
3
|
Liu F, Howard CB, Huda P, Fletcher NL, Bell CA, Blakey I, Agrez M, Thurecht KJ. Immune-modulating nanomedicines for enhanced drug delivery to non-small-cell lung cancer. Biomaterials 2025; 317:123089. [PMID: 39793167 DOI: 10.1016/j.biomaterials.2025.123089] [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: 06/11/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Immune-modulating peptides have shown potential as novel immune-stimulating agents which enhance the secretion of anticancer cytokines in vitro. However, fast clearance from blood hampers the ability of such peptides to accumulate in the tumour and results in limited therapeutic efficacy in animal studies. To address the fast blood clearance, this work reports the development and validation of a novel polymeric nanoparticle delivery system for the efficient localization of an immunomodulating peptide in the tumour microenvironment (TME). To identify the optimal polymeric nanoparticle for this study, two types of nanoparticles were developed as either branched polymers or micelles that have similar chemical functionality but different sizes. The effect of targeting the nanomedicine to the tumour-specific antigen, glycoprotein GPC-1, was explored using a bispecific antibody (BsAb) that shows an affinity for the cell protein (GPC-1) and the nanoparticle. These systems were evaluated for targeting efficiency and tumour penetration using tumour spheroids of Lewis Lung Cancer (LLC) cells and it was shown that the targeted system significantly enhanced cell association compared to the untargeted control with minor differences in penetration. The lead micelle-peptide conjugates were identified and using in vivo allograft models they were demonstrated to have high delivery efficiency of the peptide to tumours, prolonged blood circulation, enhanced tumour accumulation and tumour suppression that was associated with immune cell recruitment to the tumour.
Collapse
Affiliation(s)
- Feifei Liu
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia
| | - Christopher B Howard
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Pie Huda
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Craig A Bell
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Idriss Blakey
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Michael Agrez
- ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia; InterK Peptide Therapeutics Limited, Sydney, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging (CAI), Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, QLD, 4072, Australia; ARC Training Centre for Innovation in Biomedical Imaging Technology, University of Queensland, QLD, Australia.
| |
Collapse
|
4
|
Luo X, Wang Z, Yang M, Lei R, Yang J, Li M, Hao H, Liu Z, Tang H, Peng B, Tian Y, Chen K, Chang YN, Yuan H, Tong J, Zhu L, Lu C, Xing G, Li J. Intratumoral Delivery of High Boron-Containing Carbon Quantum Dots for Enhanced Boron Neutron Capture Therapy. ACS APPLIED BIO MATERIALS 2025. [PMID: 40392650 DOI: 10.1021/acsabm.5c00479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
The development of boron-containing carbon quantum dots for boron neutron capture therapy (BNCT) offers a promising strategy for treating refractory tumors. We synthesized carbon quantum dots with optimized parameters including feed ratio, reaction time, temperature, and pH, followed by a purification process to obtain stable, boric-acid-based boron-rich carbon quantum dots (BAQDs). The large-scale production of BAQDs was implemented by this dependable and simple synthesis method. With a boron content of 15.47 wt %, the BAQDs surpassed the clinical drug boronophenylalanine. It is also the highest recorded in BNCT-related carbon quantum dot materials research. Furthermore, BAQDs exhibited excellent water solubility, stability, and biocompatibility. The experimental results demonstrated that BAQDs exhibited an effective inhibition performance against 4T1 cells under neutron irradiation. Furthermore, BAQDs displayed successful tumor-targeting delivery capabilities. In vivo biodistribution analyses revealed efficient accumulation of BAQDs at the tumor site, with a boron content of 124.32 ± 27.64 ppm 0.5 h post-injection. With their effective boron delivery capability, BAQDs represent a promising boron delivery agent for tumor therapy via BNCT, with substantial potential for clinical application.
Collapse
Affiliation(s)
- Xianwei Luo
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Zhijie Wang
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Mingxin Yang
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Runhong Lei
- Department of Radiation Oncology, Peking University Third Hospital, Beijing 100191, China
| | - Jingru Yang
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Mengyao Li
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Haoyang Hao
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Zhicai Liu
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Hongyu Tang
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Bingbing Peng
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Youjia Tian
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Kui Chen
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Ya-Nan Chang
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Hui Yuan
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jianfei Tong
- Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Spallation Neutron Source Science Center, Dongguan 523803, China
| | - Linbo Zhu
- Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Spallation Neutron Source Science Center, Dongguan 523803, China
| | - Chengyu Lu
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Gengmei Xing
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Juan Li
- University of Chinese Academy of Sciences, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| |
Collapse
|
5
|
Jha S, Hegde M, Banerjee R, Alqahtani MS, Abbas M, Fardoun HM, Unnikrishnan J, Sethi G, Kunnumakkara AB. Nanoformulations: Reforming treatment for non-small cell lung cancer metastasis. Biochem Pharmacol 2025; 238:116928. [PMID: 40288544 DOI: 10.1016/j.bcp.2025.116928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/17/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
Non-small cell lung cancer (NSCLC) is frequently diagnosed at an advanced stage, with 20 % of cases presenting as localized disease, 25 % with regional metastasis, and 55 % with distant metastasis, contributing significantly to increased morbidity and mortality rates. Current treatments, including chemotherapy, immunotherapy, radiotherapy and targeted therapy, have shown therapeutic efficacy but are limited by issues such as lack of specificity, cytotoxicity, and therapeutic resistance. Nanoparticles (NPs) offer promising solutions to these challenges by enhancing drug penetration and retention, improving biocompatibility and stability, and achieving greater precision in targeting cancer cells. This review provides insights into various types of NPs utilized in anti-metastatic drug delivery, emphasizing their ability to enhance the efficacy of existing chemotherapeutics for the prophylaxis of metastatic NSCLC. The usage of NPs as carriers of synthetic and natural compounds aimed at inhibiting cancer cell migration and invasion have also been reviewed. Special attention has been given to biomimetic nanomaterials including extracellular vesicles and engineered exosomes, that are capable of targeting molecular pathways such as EMT, p53 and PI3K/Akt to treat metastatic NSCLC. Additionally, emphasis has been given to clinical trials of these nanoformulations and their efficacy. Although therapeutic outcomes have demonstrated certain improvements, challenges related to toxicity persist, highlighting the need for further optimization of these formulations to enhance safety and efficacy. Finally, we discuss the current limitations and future perspectives for integrating NPs into clinical settings as novel therapeutic agents for lung cancer metastasis.
Collapse
Affiliation(s)
- Shristy Jha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Mangala Hegde
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Ruchira Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Habib M Fardoun
- Research Department, Canadian University Dubai, Dubai 117781, United Arab Emirates
| | - Jyothsna Unnikrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600 Singapore.
| | - Ajaikumar B Kunnumakkara
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| |
Collapse
|
6
|
Ghazi R, Ibrahim TK, Nasir JA, Gai S, Ali G, Boukhris I, Rehman Z. Iron oxide based magnetic nanoparticles for hyperthermia, MRI and drug delivery applications: a review. RSC Adv 2025; 15:11587-11616. [PMID: 40230636 PMCID: PMC11995399 DOI: 10.1039/d5ra00728c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
Iron-oxide nanoparticles (IONPs) have garnered substantial attention in both research and technological domains due to their exceptional chemical and physical properties. These nanoparticles have mitigated the adverse effects of conventional treatment procedures by facilitating advanced theranostic approaches in integration with biomedicine. These IONPs have been extensively utilized in MRI (as contrast agents in diagnosis), drug delivery (as drug carriers), and hyperthermia (treatment), demonstrating promising results with potential for further enhancement. This study elucidates the operational principles of these NPs during diagnosis, drug delivery, and treatment, and emphasizes their precision and efficacy in transporting therapeutic agents to targeted sites without drug loss. It also analyses various challenges associated with the application of these IONPs in this field, such as biocompatibility, agglomeration, and toxicity. Furthermore, diverse strategies have been delineated to address these challenges. Overall, this review provides a comprehensive overview of the applications of IONPs in the field of biomedicine and treatment, along with the associated challenges. It offers significant assistance to researchers, professionals, and clinicians in the field of biomedicine.
Collapse
Affiliation(s)
- Rizwana Ghazi
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-(051)90642241 +92-(051)90642245
| | - Talib K Ibrahim
- Department of Petroleum Engineering, College of Engineering, Knowledge University Erbil Iraq
- Department of Petroleum Engineering, Al-Kitab University Altun Kupri Iraq
| | - Jamal Abdul Nasir
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-(051)90642241 +92-(051)90642245
| | - Shili Gai
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University Harbin 150001 P. R. China
| | - Ghafar Ali
- Nanomaterials Research Group (NRG), Physics Division, PINSTECH Nilore Islamabad Pakistan
| | - Imed Boukhris
- Department of Physics, Faculty of Science, King Khalid University P. O. Box 9004 Abha Saudi Arabia
| | - Ziaur Rehman
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-(051)90642241 +92-(051)90642245
| |
Collapse
|
7
|
Nie F, Li L, Bai Y, Yang J. Early Detection of Brain Metastases from Triple-Negative Breast Cancer with a Tumor-Targeting Dual-Modal MR/NIRF Imaging Probe. Int J Nanomedicine 2025; 20:3697-3712. [PMID: 40134526 PMCID: PMC11932937 DOI: 10.2147/ijn.s498629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Objective Imaging early-stage brain metastases from triple-negative breast cancer (TNBC) is challenging due to the blood-brain barrier (BBB). To address this issue, we developed Den-Angio-GE11, a nanoprobe engineered to traverse the BBB and selectively target metastatic cells. Methods A TNBC brain metastasis model was established in mice through intracardiac injection of MDA-MB-231 brain-seeking cells (MDA-MB-231-BR). Metastatic lesions were longitudinally monitored using T2-weighted magnetic resonance imaging (MRI) and confirmed through contrast-enhanced MRI with Gadolinium-DTPA (Gd-DTPA). The Den-Angio-GE11 nanoprobe was synthesized on a polyamidoamine (PAMAM)-G5 dendrimer platform, incorporating Angiopep-2 and GE11 peptides for BBB traversal and metastatic cell targeting. Dual-modal imaging capability was achieved by conjugating Gd-DTPA for MRI and NIR783 for near-infrared fluorescence (NIRF) imaging. Results Den-Angio-GE11 demonstrated significantly enhanced affinity to EGFR compared to controls, as confirmed by immunofluorescence staining and flow cytometry assays. Brain metastases appeared on T2-weighted MRI three weeks post-injection of MDA-MB-231BR cells and maintained uncompromised BBB function for another one or two weeks, as demonstrated by a lack of enhancement in Gd-DTPA-enhanced MRI. Compared to control nanoparticles, Den-Angio-GE11 remarkably enhanced T1 and NIRF signals of lesions after administration. Histological analysis confirmed Den-Angio-GE11 targeting brain metastatic cells. For lesions in extreme-early stage (undetectable by T2-weighted imaging), NIRF imaging post-Den-Angio-GE11 administration successfully indicated potential lesions. Fluorescence imaging analyses further verified Den-Angio-GE11 targeted sporadically metastatic cells in the brain parenchyma. Conclusion Early brain metastases of TNBC can be detected by Den-Angio-GE11 through T1-weighted MRI or NIRF imaging.
Collapse
Affiliation(s)
- Fang Nie
- Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, State Key Laboratory of Digital Medical Engineering, Zhongda Hospital, Medical School, Southeast University, Nanjing, People’s Republic of China
| | - Lin Li
- Department of Radiology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yingying Bai
- Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, State Key Laboratory of Digital Medical Engineering, Zhongda Hospital, Medical School, Southeast University, Nanjing, People’s Republic of China
| | - Jian Yang
- Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, State Key Laboratory of Digital Medical Engineering, Zhongda Hospital, Medical School, Southeast University, Nanjing, People’s Republic of China
- Department of Physiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, State Key Laboratory of Digital Medical Engineering, Zhongda Hospital, Medical School, Southeast University, Nanjing, People’s Republic of China
| |
Collapse
|
8
|
Roszkowski S, Durczyńska Z, Szablewska S. Targeted nanodelivery systems for personalized cancer therapy. Rep Pract Oncol Radiother 2025; 29:776-788. [PMID: 40104662 PMCID: PMC11912883 DOI: 10.5603/rpor.103524] [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: 11/27/2023] [Accepted: 11/12/2024] [Indexed: 03/20/2025] Open
Abstract
Conventional cancer therapies such as chemotherapy face challenges such as poor tumor targeting, systemic toxicity, and drug resistance. Nanotechnology offers solutions through advanced drug delivery systems that preferentially accumulate in tumors while avoiding healthy tissues. Recent innovations have enabled the optimization of engineered nanocarriers for extended circulation and tumor localization via both passive and active targeting mechanisms. Passive accumulation exploits the leaky vasculature of tumors, whereas active strategies use ligands to selectively bind cancer cell receptors. Multifunctional nanoparticles also allow the combination of imaging, multiple therapeutic modalities and on-demand drug release within a single platform. Overall, precisely tailored nanotherapeutics that leverage unique pathophysiological traits of malignancies provide opportunities to overcome the limitations of traditional treatment regimens. This emerging field promises more effective and personalized nanomedicine approaches to detect and treat cancer. The key aspects highlighted in this review include the biological barriers associated with nanoparticles, rational design principles to optimize nanocarrier pharmacokinetics and tumor uptake, passive and active targeting strategies, multifunctionality, and reversal of multidrug resistance.
Collapse
Affiliation(s)
- Szymon Roszkowski
- Division of Biochemistry and Biogerontology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Zofia Durczyńska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Sylwia Szablewska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| |
Collapse
|
9
|
Dey S, Ghosh M, Dev A. Signalling and molecular pathways, overexpressed receptors of colorectal cancer and effective therapeutic targeting using biogenic silver nanoparticles. Gene 2025; 936:149099. [PMID: 39557372 DOI: 10.1016/j.gene.2024.149099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Increasing morbidity and mortality in CRC is a potential threat to human health. The major challenges for better treatment outcomes are the heterogeneity of CRC cases, complicated molecular pathway cross-talks, the influence of gut dysbiosis in CRC, and the lack of multimodal target-specific drug delivery. The overexpression of many receptors in CRC cells may pave the path for targeting them with multiple ligands. The design of a more target-specific drug-delivery device with multiple ligand-functionalized, green-synthesized silver nanoparticles is highly promising and may also deliver other approved chemotherapeutic agents. This review presents the various aspects of colorectal cancer and over-expressed receptors that can be targeted with appropriate ligands to enhance the specific drug delivery potency of green synthesised silver nanoparticles. This review aims to broaden further research into this multi-ligand functionalised, safer and effective silver nano drug delivery system.
Collapse
Affiliation(s)
- Sandip Dey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India.
| |
Collapse
|
10
|
Srinivasarao DA, Shah S, Famta P, Vambhurkar G, Jain N, Pindiprolu SKSS, Sharma A, Kumar R, Padhy HP, Kumari M, Madan J, Srivastava S. Unravelling the role of tumor microenvironment responsive nanobiomaterials in spatiotemporal controlled drug delivery for lung cancer therapy. Drug Deliv Transl Res 2025; 15:407-435. [PMID: 39037533 DOI: 10.1007/s13346-024-01673-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
Design and development of efficient drug delivery technologies that impart site-specificity is the need of the hour for the effective treatment of lung cancer. The emergence of materials science and nanotechnology partially helped drug delivery scientists to achieve this objective. Various stimuli-responsive materials that undergo degradation at the pathological tumor microenvironment (TME) have been developed and explored for drug delivery applications using nanotechnological approaches. Nanoparticles (NPs), owing to their small size and high surface area to volume ratio, demonstrated enhanced cellular internalization, permeation, and retention at the tumor site. Such passive accumulation of stimuli-responsive materials helped to achieve spatiotemporally controlled and targeted drug delivery within the tumors. In this review, we discussed various stimuli-physical (interstitial pressure, temperature, and stiffness), chemical (pH, hypoxia, oxidative stress, and redox state), and biological (receptor expression, efflux transporters, immune cells, and their receptors or ligands)-that are characteristic to the TME. We mentioned an array of biomaterials-based nanoparticulate delivery systems that respond to these stimuli and control drug release at the TME. Further, we discussed nanoparticle-based combinatorial drug delivery strategies. Finally, we presented our perspectives on challenges related to scale-up, clinical translation, and regulatory approvals.
Collapse
Affiliation(s)
- Dadi A Srinivasarao
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India.
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Ganesh Vambhurkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Naitik Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Sai Kiran S S Pindiprolu
- Aditya Pharmacy College, Surampalem, 533 437, Andhra Pradesh, India
- Jawaharlal Nehru Technological University, Kakinada, 533 003, Andhra Pradesh, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Hara Prasad Padhy
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Meenu Kumari
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), 500037, Telangana, Hyderabad, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
11
|
Su X, Zhong H, Zeng Y, Zhang Y, Zhang B, Guo W, Huang Q, Ye Y. Dual-ligand-functionalized nanostructured lipid carriers as a novel dehydrocavidine delivery system for liver fibrosis therapy. Colloids Surf B Biointerfaces 2025; 246:114376. [PMID: 39551037 DOI: 10.1016/j.colsurfb.2024.114376] [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: 08/06/2024] [Revised: 10/12/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Liver fibrosis is a common stage of various chronic liver diseases, often developing into liver cirrhosis, and even liver cancer. Activated hepatic stellate cells (aHSCs) have been shown to promote the development of liver fibrosis. Therefore, dual-targeted combination therapy for liver may be an effective strategy for the treatment of liver fibrosis. PURPOSE In this study, the novel nanostructured lipid carriers (GA&GalNH2-DC-NLCs) were prepared for Dehydrocavidine (DC), glycyrrhetinic acid (GA) and galactose-PEG2000-NH2 (GalNH2) were selected as targeted ligand-modified nanostructured lipid carriers (NLCs), which enables dual-targeting to the liver for the treatment of liver fibrosis. STUDY DESIGN To study the targeting effect of GA&GalNH2-DC-NLCs on liver and its therapeutic effect on liver fibrosis, we established aHSC-T6 cell model and rat model of liver fibrosis for study. RESULTS GA&GalNH2-DC-NLCs promoted drug liver targeting efficiency and apoptosis rate by upregulating the expression of Bax. It showed that compared with no and/or GA-modified NLCs and GalNH2-modified NLCs, GA&GalNH2-DC-NLCs exhibited less extracellular matrix (ECM) deposition, induced apoptosis of aHSCs, and stronger anti-fibrosis effects in vivo. This may be due the fact that GA or GalNH2-modifified NLCs simultaneously block HSCs activation and inhibit the IL-6/STAT3 pathway. CONCLUSION GA&GalNH2-DC-NLCs is thus a potential strategy for liver fibrosis treatment.
Collapse
Affiliation(s)
- Xiaodan Su
- Department of Pharmacy, Guangxi Medical University, Nanning 530021, China.
| | - Huashuai Zhong
- Department of Pharmacy, Guangxi Medical University, Nanning 530021, China.
| | - Yongzhu Zeng
- Department of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Yuyan Zhang
- Department of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin 541199, China
| | - Wei Guo
- Department of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Qiujie Huang
- Department of Pharmacy, Guangxi University of Traditional Chinese Medicine, Nanning 530001, China.
| | - Yong Ye
- Department of Pharmacy, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Nanning 530021, China; Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Nanning 530021, China.
| |
Collapse
|
12
|
Park J, Kim D. Advanced Immunomodulatory Biomaterials for Therapeutic Applications. Adv Healthc Mater 2025; 14:e2304496. [PMID: 38716543 PMCID: PMC11834384 DOI: 10.1002/adhm.202304496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/15/2024] [Indexed: 05/22/2024]
Abstract
The multifaceted biological defense system modulating complex immune responses against pathogens and foreign materials plays a critical role in tissue homeostasis and disease progression. Recently developed biomaterials that can specifically regulate immune responses, nanoparticles, graphene, and functional hydrogels have contributed to the advancement of tissue engineering as well as disease treatment. The interaction between innate and adaptive immunity, collectively determining immune responses, can be regulated by mechanobiological recognition and adaptation of immune cells to the extracellular microenvironment. Therefore, applying immunomodulation to tissue regeneration and cancer therapy involves manipulating the properties of biomaterials by tailoring their composition in the context of the immune system. This review provides a comprehensive overview of how the physicochemical attributes of biomaterials determine immune responses, focusing on the physical properties that influence innate and adaptive immunity. This review also underscores the critical aspect of biomaterial-based immune engineering for the development of novel therapeutics and emphasizes the importance of understanding the biomaterials-mediated immunological mechanisms and their role in modulating the immune system.
Collapse
Affiliation(s)
- Ji‐Eun Park
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Dong‐Hwee Kim
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Department of Integrative Energy EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
- Biomedical Research CenterKorea Institute of Science and TechnologySeoul02792Republic of Korea
| |
Collapse
|
13
|
Penugonda S, Beesappagari P, Repollu M, Badiginchala P, Qudsiya S, Mala CUS, Gundawar R, Eranti B. Enhanced Anticancer Efficiency of Curcumin Co-Loaded Lawsone Solid Lipid Nanoparticles Against MCF-7 Breast Cancer Cell Lines: Optimization by Statistical JMP Software-Based Experimental Approach. Assay Drug Dev Technol 2025. [PMID: 39869018 DOI: 10.1089/adt.2024.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025] Open
Abstract
The present study highlighted enhancing the therapeutic effectiveness of curcumin (CUR) co-loaded lawsone (LS) through a solid lipid nanoparticles (SLNs)-based delivery system. The cetyl palmitate (CP), polyethylene glycol 400 (PEG), and probe sonication time (PS) were considered as independent variables whereas particle size and % entrapment efficiency (EE) were selected as dependent variables. The CUR-LS-SLN was developed by hot emulsification followed by probe sonication. A 23 factorial design was utilized in formulation development using JMP software version 17. Notably, the particle size and %EE of all the formulations were about 500 nm and greater than 75%, respectively. The zeta potential value was found to be -46.8 mV. From leverage plots significant and sensitive factors on particle size and %EE were identified. Contour plots led to the identification of an optimized formula whereby maintaining CP at 100 mg, PEG 400 at 6 mL, and PS at 10 min the desired particle size and %EE was achieved. TEM studies indicated the spherical shape of the particles. MTT assays of Michigan Cancer Foundation-7 (MCF-7) cells showed enhanced efficacy and greater cell inhibition of CUR-LS-SLN and combining both drugs using nanocarriers gave superior inhibition as compared with using either of the drugs evident from IC50 values of 3.7, 9.4, and 2.5 μM, respectively, for CUR, LS, and CUR-LS-SLN. The cells in the combination mostly had irregular cell walls and cell shrinkage was noted and greater cell reduction was also seen. It was found that the enhanced cytotoxicity effect of MCF-7 cells on the developed formulation was attributed to the drug's synergistic actions, more efficient nanocarrier internalizations, and sustained drug release from the formulation. Stability studies indicated that the optimized SLN was stable for 6 months.
Collapse
Affiliation(s)
- Shivarani Penugonda
- Department of Pharmaceutics, Raghavendra Institute of Pharmaceutical Education & Research - Autonomous, Anantapur, Andhra Pradesh, India
| | - Pranusha Beesappagari
- Department of Pharmaceutics, Raghavendra Institute of Pharmaceutical Education & Research - Autonomous, Anantapur, Andhra Pradesh, India
| | - Maddileti Repollu
- Department of Pharmaceutics, Raghavendra Institute of Pharmaceutical Education & Research - Autonomous, Anantapur, Andhra Pradesh, India
| | - Poojitha Badiginchala
- Department of Pharmaceutics, Raghavendra Institute of Pharmaceutical Education & Research - Autonomous, Anantapur, Andhra Pradesh, India
| | - Samreen Qudsiya
- Department of Pharmaceutics, Raghavendra Institute of Pharmaceutical Education & Research - Autonomous, Anantapur, Andhra Pradesh, India
| | - Chinni Usha Sree Mala
- Department of Pharmaceutics, Raghavendra Institute of Pharmaceutical Education & Research - Autonomous, Anantapur, Andhra Pradesh, India
| | - Ravi Gundawar
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Bhargav Eranti
- Department of Pharmaceutics, Raghavendra Institute of Pharmaceutical Education & Research - Autonomous, Anantapur, Andhra Pradesh, India
| |
Collapse
|
14
|
Razavi R, Khajouei G, Divsalar F, Dawi E, Amiri M. Recent advances on brain drug delivery via nanoparticles: alternative future materials for neuroscience applications; a review. Rev Neurosci 2025:revneuro-2024-0086. [PMID: 39829237 DOI: 10.1515/revneuro-2024-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025]
Abstract
Essentially, the blood-brain barrier (BBB) serves as a line of demarcation between neural tissues and the bloodstream. A unique and protective characteristic of the blood-brain barrier is its ability to maintain cerebral homeostasis by regulating the flux of molecules and ions. The inability to uphold proper functioning in any of these constituents leads to the disruption of this specialized multicellular arrangement, consequently fostering neuroinflammation and neurodegeneration. Recent advancements in nanomedicine have been regarded as a promising avenue for improving the delivery of drugs to the central nervous system in the modern era. A major benefit of this innovation is that it allows drugs to accumulate selectively within the cerebral area by circumventing the blood-brain barrier. Although brain-targeted nanomedicines have demonstrated impressive achievements, certain limitations in targeting specificity still exist. In this examination, we scrutinize the distinctive physical and chemical attributes of nanoparticles (NPs) contributing to their facilitation in BBB traversal. We explore the various mechanisms governing NP passage over the BBB, encompassing paracellular conveyance, mediated transport, as well as adsorptive- and receptor-mediated transcytosis. The therapeutic success of NPs for the treatment of brain tumors has been extensively investigated through the use of various categories of NPs. Among these are polymeric nanoparticles, liposomes, solid lipid nanoparticles, dendrimers, metallic nanoparticles, quantum dots, and nanogels. The potential utility of nanoparticles goes beyond their ability to transport pharmaceuticals. They can serve as adept imaging contrast agents, capable of being linked with imaging probes. This will facilitate tumor visualization, delineate lesion boundaries and margins, and monitor drug delivery and treatment response. Versatile nanoparticles can be engineered to effectively target neoplastic lesions, serving dual roles in diagnostic imaging and therapeutic interventions. Subsequently, this discourse explores the constraints associated with nanoparticles in the context of treating brain tumors.
Collapse
Affiliation(s)
- Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran
| | - Ghazal Khajouei
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| | - Fatemeh Divsalar
- Sina Hospital, Zarand Network and Health Center, 48463 Kerman University of Medical Sciences , Kerman, Iran
| | - Elmuez Dawi
- College of Humanities and Sciences, College of Humanities and Sciences, Department of Mathematics and Sciences, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
| | - Mahnaz Amiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| |
Collapse
|
15
|
Nikpour M, Karami Z, Rafieenia S, Adibifar A, Yazdani S, Saghatchi Zanjani F, Mortezazadeh T, Abdi Z, Rostamizadeh K. Inhibition of growth and lung metastasis of breast cancer by pH-responsive methotrexate/curcumin-loaded chitosan-stabilized nanoemulsions. Pharm Dev Technol 2025; 30:57-68. [PMID: 39772899 DOI: 10.1080/10837450.2024.2448335] [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: 06/04/2024] [Revised: 11/19/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Chemotherapeutic agents are widely used to combat breast cancer. However, due to their non-selective biodistribution, their usage is associated with severe adverse effects on healthy tissues. In this study, a chitosan-stabilized nanoemulsion (CSNE) was prepared for the codelivery of curcumin (CUR) and methotrexate (MTX). The mean diameter and polydispersity index of CUR-MTX-CSNEs were 194.63 ± 6.7 nm and 0.27 ± 0.06, respectively. Modifying the nanoemulsion surface with chitosan decreased the drug release at pH 7.4 compared to pH 5.8. The MTT test demonstrated that CUR-MTX-CSNEs were more successful in reducing the cell viability of 4T1 cells than both bare formulation and free drugs. Moreover, compared to the free drug-treated group, a 2.6 times reduction of the relative tumor volume was witnessed in CUR-MTX-CSNEs-receiving mice. Histopathological studies confirmed a more substantial inhibitory effect on tumor growth and pulmonary metastasis of developed nanostructures than free CUR/MTX. While there was no noticeable toxicity in the vital organs of CUR-MTX-CSNEs-receiving mice, free drugs resulted in severe toxicity in the liver, kidney, lung and spleen. Overall, the pH-dependent drug release, improved anti-tumor activity and reduced organ toxicity suggest that CUR-MTX-CSNE may be promising in breast cancer therapy.
Collapse
Affiliation(s)
- Mehrnoosh Nikpour
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Zahra Karami
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samaneh Rafieenia
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Arghavan Adibifar
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Shaghayegh Yazdani
- Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Tohid Mortezazadeh
- Department of Medical Physics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Abdi
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kobra Rostamizadeh
- Department of Psychiatry and Behavioral Sciences, Department of Pharmacology, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
16
|
Sayed ZS, Hieba EM, Batakoushy HA, Rashdan HRM, Ismail E, Elkatlawy SM, Elzwawy A. Cancer treatment approaches within the frame of hyperthermia, drug delivery systems, and biosensors: concepts and future potentials. RSC Adv 2024; 14:39297-39324. [PMID: 39670162 PMCID: PMC11635600 DOI: 10.1039/d4ra06992g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
This work presents a review of the therapeutic modalities and approaches for cancer treatment. A brief overview of the traditional treatment routes is presented in the introduction together with their reported side effects. A combination of the traditional approaches was reported to demonstrate an effective therapy until a few decades ago. With the improvement in the fabrication of nanomaterials, targeted therapy represents a novel therapeutic approach. This improvement established on nanoparticles is categorized into hyperthermia, drug delivery systems, and biosensors. Hyperthermia presents a personalized medicine-based approach in which targeted zones are heated up until the diseased tissue is destroyed by the thermal effect. The use of magnetic nanoparticles further improved the effectiveness of hyperthermia owing to the enhanced heating action, further increasing the accuracy of the targeting process. Nanoparticle-based biosensors present a smart nanodevice that can detect, monitor, and target tumor tissues by following the biomarkers in the body fluids. Magnetic nanoparticles offer a controlled thermo-responsive device that can be manipulated by changing the magnetic field, offering a more personalized and controlled hyperthermia therapeutic modality. Similarly, gold nanoparticles offer an effective aid in the hyperthermia treatment approach. Furthermore, carbon nanotubes and metal-organic frameworks present a cutting-edge approach to cancer treatment. A combination of functionalized nanoparticles offers a unique route for drug delivery systems, in which therapeutic agents carried by nanoparticles are guided into the human body and then released in the target spot.
Collapse
Affiliation(s)
- Zeinab S Sayed
- Faculty of Applied Medical Science, Misr University for Science and Technology (MUST) Giza Egypt
| | - Eman M Hieba
- Chemistry and Entomology Department, Faculty of Science, Cairo University Giza 12613 Egypt
| | - Hany A Batakoushy
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Menoufia University Shebin Elkom 32511 Egypt
| | - Huda R M Rashdan
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre 33 El Buhouth St., Dokki Giza 12622 Egypt
| | - Enas Ismail
- Department of Prosthodontics, Faculty of Dentistry, University of the Western Cape Cape Town 7505 South Africa
- Physics Department, Faculty of Science (Girl's Branch), Al Azhar University Nasr City 11884 Cairo Egypt
| | - Saeid M Elkatlawy
- Department of Physics, Faculty of Science, University of Sadat City Fifth Zone Sadat Egypt
| | - Amir Elzwawy
- Ceramics Department, Advanced Materials Technology and Mineral Resources Research Institute, National Research Centre (NRC) 33 El Bohouth St., Dokki Giza 12622 Egypt
| |
Collapse
|
17
|
Baena JC, Pérez LM, Toro-Pedroza A, Kitawaki T, Loukanov A. CAR T Cell Nanosymbionts: Revealing the Boundless Potential of a New Dyad. Int J Mol Sci 2024; 25:13157. [PMID: 39684867 DOI: 10.3390/ijms252313157] [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: 09/01/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer treatment has traditionally focused on eliminating tumor cells but faces challenges such as resistance and toxicity. A promising direction involves targeting the tumor microenvironment using CAR T cell immunotherapy, which has shown potential for treating relapsed and refractory cancers but is limited by high costs, resistance, and toxicity, especially in solid tumors. The integration of nanotechnology into ICAM cell therapy, a concept we have named "CAR T nanosymbiosis", offers new opportunities to overcome these challenges. Nanomaterials can enhance CAR T cell delivery, manufacturing, activity modulation, and targeting of the tumor microenvironment, providing better control and precision. This approach aims to improve the efficacy of CAR T cells against solid tumors, reduce associated toxicities, and ultimately enhance patient outcomes. Several studies have shown promising results, and developing this therapy further is essential for increasing its accessibility and effectiveness. Our "addition by subtraction model" synthesizes these multifaceted elements into a unified strategy to advance cancer treatment paradigms.
Collapse
Affiliation(s)
- Juan C Baena
- Division of Oncology, Department of Medicine, Fundación Valle del Lili, ICESI University, Carrera 98 No. 18-49, Cali 760032, Colombia
- LiliCAR-T Group, Fundación Valle del Lili, ICESI University, Cali 760032, Colombia
| | - Lucy M Pérez
- Division of Oncology, Department of Medicine, Fundación Valle del Lili, ICESI University, Carrera 98 No. 18-49, Cali 760032, Colombia
- LiliCAR-T Group, Fundación Valle del Lili, ICESI University, Cali 760032, Colombia
| | - Alejandro Toro-Pedroza
- Division of Oncology, Department of Medicine, Fundación Valle del Lili, ICESI University, Carrera 98 No. 18-49, Cali 760032, Colombia
- LiliCAR-T Group, Fundación Valle del Lili, ICESI University, Cali 760032, Colombia
| | - Toshio Kitawaki
- Department of Hematology, Kyoto University Hospital, Kyoto 606-8507, Japan
| | - Alexandre Loukanov
- Department of Chemistry and Materials Science, National Institute of Technology, Gunma College, Maebashi 371-8530, Japan
- Laboratory of Engineering Nanobiotechnology, University of Mining and Geology "St. Ivan Rilski", 1700 Sofia, Bulgaria
| |
Collapse
|
18
|
Chen F, Xue Y, Zhang W, Zhou H, Zhou Z, Chen T, YinWang E, Li H, Ye Z, Gao J, Wang S. The role of mitochondria in tumor metastasis and advances in mitochondria-targeted cancer therapy. Cancer Metastasis Rev 2024; 43:1419-1443. [PMID: 39307891 PMCID: PMC11554835 DOI: 10.1007/s10555-024-10211-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/08/2024] [Indexed: 11/05/2024]
Abstract
Mitochondria are central actors in diverse physiological phenomena ranging from energy metabolism to stress signaling and immune modulation. Accumulating scientific evidence points to the critical involvement of specific mitochondrial-associated events, including mitochondrial quality control, intercellular mitochondrial transfer, and mitochondrial genetics, in potentiating the metastatic cascade of neoplastic cells. Furthermore, numerous recent studies have consistently emphasized the highly significant role mitochondria play in coordinating the regulation of tumor-infiltrating immune cells and immunotherapeutic interventions. This review provides a comprehensive and rigorous scholarly investigation of this subject matter, exploring the intricate mechanisms by which mitochondria contribute to tumor metastasis and examining the progress of mitochondria-targeted cancer therapies.
Collapse
Affiliation(s)
- Fanglu Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yucheng Xue
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wenkan Zhang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hao Zhou
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhiyi Zhou
- The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Eloy YinWang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
19
|
Keshavarz Shahbaz S, Koushki K, Izadi O, Penson PE, Sukhorukov VN, Kesharwani P, Sahebkar A. Advancements in curcumin-loaded PLGA nanoparticle delivery systems: progressive strategies in cancer therapy. J Drug Target 2024; 32:1207-1232. [PMID: 39106154 DOI: 10.1080/1061186x.2024.2389892] [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: 04/23/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
Cancer is a leading cause of death worldwide, and imposes a substantial socioeconomic burden with little impact especially on aggressive types of cancer. Conventional therapies have many serious side effects including generalised systemic toxicity which limits their long-term use. Tumour resistance and recurrence is another main problem associated with conventional therapy. Purified or extracted natural products have been investigated as cost-effective cancer chemoprotective agents with the potential to reverse or delaying carcinogenesis. Curcumin (CUR) as a natural polyphenolic component, exhibits many pharmacological activities such as anti-cancer, anti-inflammatory, anti-microbial, activity against neurodegenerative diseases including Alzheimer, antidiabetic activities (type II diabetes), anticoagulant properties, wound healing effects in both preclinical and clinical studies. Despite these effective protective properties, CUR has several limitations, including poor aqueous solubility, low bioavailability, chemical instability, rapid metabolism and a short half-life time. To overcome the pharmaceutical problems associated with free CUR, novel nanomedicine strategies (including polymeric nanoparticles (NPs) such as poly (lactic-co-glycolic acid) (PLGA) NPs have been developed. These formulations have the potential to improve the therapeutic efficacy of curcuminoids. In this review, we comprehensively summarise and discuss recent in vitro and in vivo studies to explore the pharmaceutical significance and clinical benefits of PLGA-NPs delivery system to improve the efficacy of CUR in the treatment of cancer.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Khadijeh Koushki
- Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Omid Izadi
- Department of Industrial Engineering, ACECR Institute of Higher Education Kermanshah, Kermanshah, Iran
| | - Peter E Penson
- Clinical Pharmacy and Therapeutics Research Group, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
- Liverpool Centre for Cardiovascular Science, Liverpool, UK
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - 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 Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Alidaei-Sharif H, Babazadeh-Mamaqani M, Mohammadi-Jorjafki M, Roghani-Mamaqani H, Salami-Kalajahi M. Multi-Responsive Polymer Nanoparticles: A Versatile Platform for Double-Security Anticounterfeiting and Smart Food Packaging. Macromol Rapid Commun 2024; 45:e2400561. [PMID: 39461898 DOI: 10.1002/marc.202400561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/05/2024] [Indexed: 10/29/2024]
Abstract
Potential applications of colloidal polymer nanoparticles in the preparation of smart inks are investigated by physical incorporation of the oxazolidine molecules. Precise adjusting the polymer chain flexibility and polarity is achieved by controlling the ratio of methyl methacrylate and butyl acrylate monomers in the polymerization reaction. In addition, nanofibrous indicators of acid-base vapors are prepared from the latex nanoparticles. This can be beneficial for creating materials that sense and respond to environmental changes, such as humidity or moisture and acidity. Thermochromic inks are prepared by microencapsulating crystal violet lactone dye (CVL) in polymer matrices to prevent their release into the aqueous media. Combining two distinct systems with varying triggers, such as light and temperature, provides an effective strategy for double-encryption anticounterfeiting and crack and scratch detection and indication applications. Preparing labels impregnated with double-responsive inks, a novel approach is developed for food spoilage detection and preservation indication. Labels are manufactured using polymer nanoparticles, which contain photoluminescent oxazolidine molecules, as well as a trinary mixture of CVL within core-shell latex particles as the thermochromic dye. The combination of these two responsive elements transforms traditional packaging into a dynamic and interactive sentinel for the food it holds.
Collapse
Affiliation(s)
- Hossein Alidaei-Sharif
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, P.O. Box 51335-1996, Iran
| | - Milad Babazadeh-Mamaqani
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, P.O. Box 51335-1996, Iran
| | - Moein Mohammadi-Jorjafki
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, P.O. Box 51335-1996, Iran
| | - Hossein Roghani-Mamaqani
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, P.O. Box 51335-1996, Iran
- Institute of Polymeric Materials, Sahand University of Technology, Tabriz, P.O. Box 51335-1996, Iran
| | - Mehdi Salami-Kalajahi
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, P.O. Box 51335-1996, Iran
- Institute of Polymeric Materials, Sahand University of Technology, Tabriz, P.O. Box 51335-1996, Iran
| |
Collapse
|
21
|
Pant A, Laliwala A, Holstein SA, Mohs AM. Recent advances in targeted drug delivery systems for multiple myeloma. J Control Release 2024; 376:215-230. [PMID: 39384153 PMCID: PMC11611669 DOI: 10.1016/j.jconrel.2024.10.003] [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: 08/09/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Despite significant therapeutic advances, multiple myeloma (MM) remains a challenging, incurable, hematological malignancy. The efficacy of traditional chemotherapy and currently available anti-MM agents is in part limited by their adverse effects, which restrict their therapeutic potential. Nanotherapeutics is an emerging field of cancer therapy that can overcome the biological and chemical barriers of existing anticancer drugs. This review presents an overview of recent advancements in nanoparticle- and immunotherapy-based drug delivery systems for MM treatment. It further delves into the targeting strategies, mechanism of controlled drug release, and challenges associated with the development of drug delivery systems for the treatment of MM.
Collapse
Affiliation(s)
- Ashruti Pant
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Aayushi Laliwala
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Sarah A Holstein
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Department of Internal Medicine, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Aaron M Mohs
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, S 45th St, Omaha, NE 68198, USA.
| |
Collapse
|
22
|
Maryam S, Krukiewicz K. Sweeten the pill: Multi-faceted polysaccharide-based carriers for colorectal cancer treatment. Int J Biol Macromol 2024; 282:136696. [PMID: 39437958 DOI: 10.1016/j.ijbiomac.2024.136696] [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: 07/05/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Colorectal cancer (CRC) ranks as the second deadliest cancer globally and the third most common malignant tumor. While surgery remains the primary treatment for CRC, alternative therapies such as chemotherapy, molecular targeted therapy, and immunotherapy are also commonly used. The significant side effects and toxicity of conventional drugs drive the search for novel targeted therapies, including the design of advanced drug delivery systems. Polysaccharide-based biopolymers, with their low toxicity, non-immunogenic behavior, synergistic interactions with other biopolymers, and tissue and cell compatibility, emerge as excellent drug carriers for this application. This review aims to provide an in-depth overview of recent advancements in developing polysaccharide-based biopolymeric carriers for anticancer compounds in the treatment of CRC. We highlight the multifunctional nature of polysaccharides, showcasing their potential as standalone drug carriers or as integral components of intelligent robotic devices for biomedical therapeutic applications. In addition to exploring the opportunities for using carbohydrate polymers in CRC treatment, we address the challenges and failures that may limit their applicability in biomedical research, as well as summarize the recent preclinical and clinical trials, resulting in several commercialization attempts. This comprehensive overview critically summarizes the potential of polysaccharide-based biomaterials in CRC treatment.
Collapse
Affiliation(s)
- Sajida Maryam
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Gliwice, Poland; Joint Doctoral School, Silesian University of Technology, Gliwice, Poland
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Gliwice, Poland.
| |
Collapse
|
23
|
Cullion K, Ostertag-Hill CA, Tang W, Pan M, Kohane DS. Size-dependent Nanoparticle Accumulation In Venous Malformations. JOURNAL OF VASCULAR ANOMALIES 2024; 5:e00103. [PMID: 39734473 PMCID: PMC11670902 DOI: 10.1097/jova.0000000000000103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/22/2024] [Indexed: 12/31/2024]
Abstract
Objective The current treatment of venous malformations (VMs) consists of medications with systemic toxicity and procedural interventions with high technical difficulty and risk of hemorrhage. Using nanoparticles (NPs) to enhance drug delivery to VMs could enhance efficacy and decrease systemic toxicity. NPs can accumulate in tissues with abnormal vasculature, a concept known as the enhanced permeation and retention (EPR) effect. EPR has been documented in tumors, bioengineered vessels, and VMs. However, in VMs, it is unknown if NP size affects EPR and if so, which particle size improves NP accumulation. Methods In this study, we used a murine model of subcutaneous VMs using human umbilical vein endothelial cells that express the most frequent VM-causing tyrosine kinase with immunoglobulin and EGF homology domains mutation, tyrosine kinase with immunoglobulin and EGF homology domains-L914F. Hollow silica NPs coated in polyethylene glycol (PEG) and conjugated to a fluorophore were administered systemically via tail vein injection. We studied the accumulation of a range of NP sizes within the VM and organs using confocal microscopy and an in vivo imaging system. Results The 20, 50, 80, and 180 nm PEGylated, fluorophore-tagged hollow silica NPs were spherical and had hydrodynamic diameters of 31.6 ± 0.9, 58.5 ± 0.1, 87.1 ± 2.4, and 232 ± 1.26 nm, respectively. Following systemic NP administration, 20 nm NPs had 2 times more fluorescence accumulation within VMs compared with 50 nm, and 6 times more fluorescence accumulation compared with larger (greater than 80 nm) NPs (P < .01). Conclusion This study helps to determine the optimal NP size for passive accumulation within VMs and lays the foundation for engineering NPs for the treatment of VMs.
Collapse
Affiliation(s)
- Kathleen Cullion
- Laboratory for Biomaterials and Drug Delivery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medical Critical Care, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Claire A. Ostertag-Hill
- Laboratory for Biomaterials and Drug Delivery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Weimin Tang
- Laboratory for Biomaterials and Drug Delivery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medical Critical Care, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michelle Pan
- Laboratory for Biomaterials and Drug Delivery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medical Critical Care, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel S. Kohane
- Laboratory for Biomaterials and Drug Delivery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Anesthesiology, Critical Care, and Pain Management, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
24
|
Chao PH, Chan V, Li SD. Nanomedicines modulate the tumor immune microenvironment for cancer therapy. Expert Opin Drug Deliv 2024; 21:1719-1733. [PMID: 39354745 DOI: 10.1080/17425247.2024.2412245] [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: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/03/2024]
Abstract
INTRODUCTION In recent years, the evolution of immunotherapy as a means to trigger a robust antitumor immune response has revolutionized cancer treatment. Despite its potential, the effectiveness of cancer immunotherapy is hindered by low response rates and significant systemic side effects. Nanotechnology emerges as a promising frontier in shaping the future of cancer immunotherapy. AREAS COVERED This review elucidates the pivotal role of nanomedicine in reshaping the immune tumor microenvironment and explores innovative strategies pursued by diverse research groups to enhance the therapeutic efficacy of cancer immunotherapy. It discusses the hurdles encountered in cancer immunotherapy and the application of nanomedicine for small molecule immune modulators and nucleic acid therapeutics. It also highlights the advancements in DNA and mRNA vaccines facilitated by nanotechnology and outlines future trajectories in this evolving field. EXPERT OPINION Collectively, the integration of nanomedicine into cancer immunotherapy stands as a promising avenue to tackle the intricacies of the immune tumor microenvironment. Innovations such as immune checkpoint inhibitors and cancer vaccines have shown promise. Future developments will likely optimize nanoparticle design through artificial intelligence and create biocompatible, multifunctional nanoparticles, promising more effective, personalized, and durable cancer treatments, potentially transforming the field in the foreseeable future.
Collapse
Affiliation(s)
- Po-Han Chao
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Vanessa Chan
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Liu L, Yang M, Chen Z. Surface functionalized nanomaterial systems for targeted therapy of endocrine related tumors: a review of recent advancements. Drug Deliv 2024; 31:2390022. [PMID: 39138394 PMCID: PMC11328606 DOI: 10.1080/10717544.2024.2390022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
The application of multidisciplinary techniques in the management of endocrine-related cancers is crucial for harnessing the advantages of multiple disciplines and their coordinated efforts in eliminating tumors. Due to the malignant characteristics of cancer cells, they possess the capacity to develop resistance to traditional treatments such as chemotherapy and radiotherapy. Nevertheless, despite diligent endeavors to enhance the prediction of outcomes, the overall survival rate for individuals afflicted with endocrine-related malignancy remains quite miserable. Hence, it is imperative to investigate innovative therapy strategies. The latest advancements in therapeutic tactics have offered novel approaches for the therapy of various endocrine tumors. This paper examines the advancements in nano-drug delivery techniques and the utilization of nanomaterials for precise cancer cures through targeted therapy. This review provides a thorough analysis of the potential of combined drug delivery strategies in the treatment of thyroid cancer, adrenal gland tumors, and pancreatic cancer. The objective of this study is to gain a deeper understanding of current therapeutic approaches, stimulate the development of new drug DDS, and improve the effectiveness of treatment for patients with these diseases. The intracellular uptake of pharmaceuticals into cancer cells can be significantly improved through the implantation of synthetic or natural substances into nanoparticles, resulting in a substantial reduction in the development of endocrine malignancies.
Collapse
Affiliation(s)
- Limei Liu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Miao Yang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ziyang Chen
- Department of Gastroenterology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
26
|
Sorroza-Martínez K, González-Sánchez I, Villamil-Ramos R, Cerbón M, Guerrero-Álvarez JA, Coronel-Cruz C, Rivera E, González-Méndez I. Using Poly(amidoamine) PAMAM-βCD Dendrimer for Controlled and Prolonged Delivery of Doxorubicin as Alternative System for Cancer Treatment. Pharmaceutics 2024; 16:1509. [PMID: 39771488 PMCID: PMC11728618 DOI: 10.3390/pharmaceutics16121509] [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: 10/08/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: Doxorubicin (Dox) is an anticancer drug used in the treatment of a wide range of solid tumors; however, Dox causes systemic toxicity and irreversible cardiotoxicity. The design of a new nanosystem that allows for the control of Dox loading and delivery results is a powerful tool to control Dox release only in cancer cells. For this reason, supramolecular self-assembly was performed between a poly(amidoamine) (PAMAM) dendrimer decorated with four β-cyclodextrin (βCD) units (PAMAM-βCD) and an adamantane-hydrazone-doxorubicin (Ad-h-Dox) prodrug. Methods: The formation of inclusion complexes (ICs) between the prodrug and all the βCD cavities present on the surface of the PAMAM-βCD dendrimer was followed by 1H-NMR titration and corroborated by 2D NOESY experiments. A full characterization of the supramolecular assembly was performed in the solid state by thermal analysis (DSC/TGA) and scanning electron microscopy (SEM) and in solution by the DOSY NMR technique in D2O. Furthermore, the Dox release profiles from the PAMAM-βCD/Ad-h-Dox assembly at different pH values was studied by comparing the efficiency against a native βCD/Ad-h-Dox IC. Additionally, in vitro cytotoxic activity assays were performed for the nanocarrier alone and the two supramolecular assemblies in different carcinogenic cell lines. Results: The PAMAM-βCD/Ad-h-Dox assembly was adequately characterized, and the cytotoxic activity results demonstrate that the nanocarrier alone and its hydrolysis product are innocuous compared to the PAMAM-βCD/Ad-h-Dox nanocarrier that showed cytotoxicity equivalent to free Dox in the tested cancer cell lines. The in vitro drug release assays for the PAMAM-βCD/Ad-h-Dox system showed an acidic pH-dependent behavior and a prolonged profile of up to more than 72 h. Conclusions: The design of PAMAM-βCD/Ad-h-Dox consists of a new controlled and prolonged Dox release system for potential use in cancer treatment.
Collapse
Affiliation(s)
- Kendra Sorroza-Martínez
- Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Calzada del Hueso 1100, Col. Villa Quietud, Mexico City CP 04960, Mexico;
| | - Ignacio González-Sánchez
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Circuito Escolar, Ciudad Universitaria, Mexico City CP 04510, Mexico; (I.G.-S.); (M.C.)
| | - Raúl Villamil-Ramos
- Centro de Investigaciones Químicas, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, Cuernavaca CP 62209, Mexico; (R.V.-R.); (J.A.G.-Á.)
| | - Marco Cerbón
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Circuito Escolar, Ciudad Universitaria, Mexico City CP 04510, Mexico; (I.G.-S.); (M.C.)
| | - Jorge Antonio Guerrero-Álvarez
- Centro de Investigaciones Químicas, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, Cuernavaca CP 62209, Mexico; (R.V.-R.); (J.A.G.-Á.)
| | - Cristina Coronel-Cruz
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Circuito Escolar, Ciudad Universitaria, Mexico City CP 04510, Mexico;
| | - Ernesto Rivera
- Departamento de Reología, Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Mexico City CP 04510, Mexico
| | - Israel González-Méndez
- Centro de Investigaciones Químicas, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, Cuernavaca CP 62209, Mexico; (R.V.-R.); (J.A.G.-Á.)
| |
Collapse
|
27
|
Shi S, Zhong H, Zhang Y, Mei Q. Targeted delivery of nano-radiosensitizers for tumor radiotherapy. Coord Chem Rev 2024; 518:216101. [DOI: 10.1016/j.ccr.2024.216101] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
Glickstein B, Shaul O, Ilovitsh T. Rationally Designed Acoustic Holograms for Uniform Nanodroplet-Mediated Tissue Ablation. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2024; 71:1606-1615. [PMID: 39352816 DOI: 10.1109/tuffc.2024.3471873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Nanodroplets (NDs) are phase-changing agents that have shown great potential for ultrasound (US) applications. When US is applied, NDs can undergo a phase transition into gas bubbles, enabling cavitation that can be used to reduce the pressure threshold required for mechanical ablation of tissues. Effective tissue fractionation depends on precise vaporization to achieve uniform and predictable bubble formation. This study aimed to optimize ND vaporization using acoustic holograms for improved ND-mediated histotripsy. Tissue ablation was conducted using a two-step approach, where a rotating imaging probe was used for ND vaporization followed by low-frequency US for detonation. We developed and validated three distinct acoustic hologram patterns targeting different regions within a circular area through simulations and experiments. Using custom-made gelatin phantoms designed for optimal ND vaporization imaging, the superpositioned patterns demonstrated significantly more uniform ND vaporization compared to standard single-focus steering, with ND coverage reaching % for the optimized vaporization approach versus % for the single focus steering. Ex vivo chicken liver experiments confirmed the enhanced efficiency of the optimized approach, resulting in significantly larger and more uniform lesion areas. Lesion areas generated by 120 s of treatment reached mm2 compared to mm2 for the standard approach, a 5.1-fold increase. These findings suggest that using acoustic holograms can improve ND vaporization uniformity and enhance the homogeneity of tissue fractionation, thereby potentially enhancing therapeutic outcomes.
Collapse
|
29
|
Chatterjee S, Sil PC. Mechanistic Insights into Toxicity of Titanium Dioxide Nanoparticles at the Micro- and Macro-levels. Chem Res Toxicol 2024; 37:1612-1633. [PMID: 39324438 DOI: 10.1021/acs.chemrestox.4c00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Titanium oxide nanoparticles (TiO2 NPs) have been regarded as a legacy nanomaterial due to their widespread usage across multiple fields. The TiO2 NPs have been and are still extensively used as a food and cosmetic additive and in wastewater and sewage treatment, paints, and industrial catalysis as ultrafine TiO2. Recent developments in nanotechnology have catapulted it into a potent antibacterial and anticancer agent due to its excellent photocatalytic potential that generates substantial amounts of highly reactive oxygen radicals. The method of production, surface modifications, and especially size impact its toxicity in biological systems. The anatase form of TiO2 (<30 nm) has been found to exert better and more potent cytotoxicity in bacteria as well as cancer cells than other forms. However, owing to the very small size, anatase particles are able to penetrate deep tissue easily; hence, they have also been implicated in inflammatory reactions and even as a potent oncogenic substance. Additionally, TiO2 NPs have been investigated to assess their toxicity to large-scale ecosystems owing to their excellent reactive oxygen species (ROS)-generating potential compounded with widespread usage over decades. This review discusses in detail the mechanisms by which TiO2 NPs induce toxic effects on microorganisms, including bacteria and fungi, as well as in cancer cells. It also attempts to shed light on how and why it is so prevalent in our lives and by what mechanisms it could potentially affect the environment on a larger scale.
Collapse
Affiliation(s)
- Sharmistha Chatterjee
- Division of Molecular Medicine, Bose Institute, P 1/12, CIT Scheme VIIM, Kankurgachi, Kolkata-700054, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P 1/12, CIT Scheme VIIM, Kankurgachi, Kolkata-700054, India
| |
Collapse
|
30
|
Li J, Li B, Liu F, Deng M, Zhang Z, Ran Y, Wang B. A multifunctional nanosystem catalyzed by cascading natural glucose oxidase and Fe 3O 4 nanozymes for synergistic chemodynamic and photodynamic cancer therapy. Acta Biomater 2024:S1742-7061(24)00617-2. [PMID: 39426656 DOI: 10.1016/j.actbio.2024.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
The significance of the tumor microenvironment (TME) in tumor initiation and progression is increasingly acknowledged. Conventional therapeutic approaches face limitations within the complex TME, including the restrictions imposed by hypoxia on photodynamic therapy (PDT) and the deficiency of endogenous H₂O₂ affecting chemodynamic therapy (CDT). In response to the TME's characteristics of high metabolism, hypoxia, and weak acidity, a multifunctional nanosystem MNPs/GOD@CS/IR820, which synergistically integrates CDT and PDT, has been developed. This system can actively accumulate at tumor sites under an external magnetic field and release active components in response to the weakly acidic TME. It mitigates the limitations imposed by hypoxia and endogenous H₂O₂ deficiency on PDT and CDT, respectively, thereby enabling synergistic treatment. Additionally, the system's multimodal imaging capabilities facilitate precise tumor localization and real-time, non-invasive in vivo assessment via fluorescence imaging and MRI. In vitro and in vivo evaluations demonstrate significant antitumor efficacy, effectively inhibiting tumor growth and improving survival rates. By comprehensively addressing the challenges posed by the complex TME and enhancing real-time monitoring capabilities, our nanosystem paves the way for personalized and precise cancer treatment. STATEMENT OF SIGNIFICANCE: This study introduces an innovative MNPs/GOD@CS/IR820 nanosystem that represents a significant advancement in cancer nanomedicine by addressing critical limitations of conventional photodynamic therapy (PDT), particularly in hypoxic tumor microenvironments. By synergistically integrating chemodynamic therapy (CDT) with PDT and incorporating MRI and fluorescence dual-modal imaging capabilities, this multifunctional platform offers enhanced therapeutic efficacy and real-time monitoring. The system's ability to generate oxygen in situ overcomes hypoxia-induced limitations, while its multimodal mechanism of action induces tumor cell apoptosis through multiple pathways. In vitro and in vivo studies demonstrate remarkable antitumor efficacy across diverse cancer types, significantly inhibiting tumor growth and improving survival rates. This comprehensive approach to cancer diagnosis and treatment not only advances precision medicine for targeted, multimodal cancer management but also provides a promising foundation for future clinical applications, potentially transforming cancer treatment strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Jiale Li
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 30018, China
| | - Bo Li
- Department of Radiology, West China Hospital, Sichuan University, Wuhou District, Chengdu City, Sichuan 610041, China
| | - Feng Liu
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 30018, China
| | - Ming Deng
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 30018, China
| | - Ziying Zhang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 30018, China
| | - Yutao Ran
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 30018, China
| | - Bing Wang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 30018, China.
| |
Collapse
|
31
|
Verma VS, Pandey A, Jha AK, Badwaik HKR, Alexander A, Ajazuddin. Polyethylene Glycol-Based Polymer-Drug Conjugates: Novel Design and Synthesis Strategies for Enhanced Therapeutic Efficacy and Targeted Drug Delivery. Appl Biochem Biotechnol 2024; 196:7325-7361. [PMID: 38519751 DOI: 10.1007/s12010-024-04895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 03/25/2024]
Abstract
Due to their potential to enhance therapeutic results and enable targeted drug administration, polymer-drug conjugates that use polyethylene glycol (PEG) as both the polymer and the linker for drug conjugation have attracted much research. This study seeks to investigate recent developments in the design and synthesis of PEG-based polymer-drug conjugates, emphasizing fresh ideas that fill in existing knowledge gaps and satisfy the increasing need for more potent drug delivery methods. Through an extensive review of the existing literature, this study identifies key challenges and proposes innovative strategies for future investigations. The paper presents a comprehensive framework for designing and synthesizing PEG-based polymer-drug conjugates, including rational molecular design, linker selection, conjugation methods, and characterization techniques. To further emphasize the importance and adaptability of PEG-based polymer-drug conjugates, prospective applications are highlighted, including cancer treatment, infectious disorders, and chronic ailments.
Collapse
Affiliation(s)
- Vinay Sagar Verma
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India
| | - Aakansha Pandey
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Arvind Kumar Jha
- Shri Shankaracharya Professional University, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Hemant Kumar Ramchandra Badwaik
- Shri Shankaracharya College of Pharmaceutical Sciences, Junwani, Bhilai, 490020, Chhattisgarh, India.
- Shri Shankaracharya Institute of Pharmaceutical Sciences and Research, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India.
| | - Amit Alexander
- Department of Pharmaceuticals, National Institute of Pharmaceutical Education and Research, Ministry of Chemical and Fertilizers, Guwahati, 781101, Assam, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India.
| |
Collapse
|
32
|
López-Espinosa J, Park P, Holcomb M, Godin B, Villapol S. Nanotechnology-driven therapies for neurodegenerative diseases: a comprehensive review. Ther Deliv 2024; 15:997-1024. [PMID: 39297726 PMCID: PMC11583628 DOI: 10.1080/20415990.2024.2401307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 11/22/2024] Open
Abstract
Neurological diseases, characterized by neuroinflammation and neurodegeneration, impose a significant global burden, contributing to substantial morbidity, disability and mortality. A common feature of these disorders, including stroke, traumatic brain injury and Alzheimer's disease, is the impairment of the blood-brain barrier (BBB), a critical structure for maintaining brain homeostasis. The compromised BBB in neurodegenerative conditions poses a significant challenge for effective treatment, as it allows harmful substances to accumulate in the brain. Nanomedicine offers a promising approach to overcoming this barrier, with nanoparticles (NPs) engineered to deliver therapeutic agents directly to affected brain regions. This review explores the classification and design of NPs, divided into organic and inorganic categories and further categorized based on their chemical and physical properties. These characteristics influence the ability of NPs to carry and release therapeutic agents, target specific tissues and ensure appropriate clearance from the body. The review emphasizes the potential of NPs to enhance the diagnosis and treatment of neurodegenerative diseases through targeted delivery, improved drug bioavailability and real-time therapeutic efficacy monitoring. By addressing the challenges of the compromised BBB and targeting inflammatory biomarkers, NPs represent a cutting-edge strategy in managing neurological disorders, promising better patient outcomes.
Collapse
Affiliation(s)
- Jessica López-Espinosa
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
- School of Medicine and Health Sciences of Tecnológico de Monterrey, Guadalajara, México
| | - Peter Park
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
| | - Morgan Holcomb
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TXUSA
- Department of Obstetrics & Gynecology, Houston Methodist Hospital, Houston, TXUSA
- Department of Obstetrics & Gynecology, Weill Cornell Medicine College, New York, NYUSA
- Department of Biomedical Engineering, Texas A&M University, College Station, TXUSA
| | - Sonia Villapol
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, New York, NY USA
| |
Collapse
|
33
|
Bhargav E, Mohammed N, Singh UN, Ramalingam P, Challa RR, Vallamkonda B, Ahmad SF, Dsnbk P, Pasala PK, Rudrapal M. A central composite design-based targeted quercetin nanoliposomal formulation: Optimization and cytotoxic studies on MCF-7 breast cancer cell lines. Heliyon 2024; 10:e37430. [PMID: 39296160 PMCID: PMC11409131 DOI: 10.1016/j.heliyon.2024.e37430] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/21/2024] Open
Abstract
This study aimed to enhance the efficacy of quercetin (QT) by formulating it into a liposomal drug delivery system utilizing the concept of central composite design. The drug:lipid ratio, cholesterol concentration, and sonication time were selected as independent variables in the study. The vesicle and percentage entrapment efficiency were selected as the dependent variables. Quercetin nanoliposomes (QT-NLs) were prepared via a combination of ethanol injection and thin film hydration. The vesicle size and entrapment efficiency of all formulations were within the ranges of 100 nm and >80 %, respectively. The zeta potential value indicated the stability of the optimized formulation. The contour plots were used to select the desired batch range. SEM studies revealed an imperfect crystalline morphology without any unwanted agglomeration. MTT assays on VERO cell lines indicated the safety of the developed formulation. MTT assays of MCF-7 cells revealed IC50 values of 5.8 μM and 7.9 μM for QT-NLs and QT, respectively. In our study, the optimized formulation exhibited late and early apoptosis and necrosis when used to treat MCF-7 cells. S and G2/M cell cycle phases of MCF-7 cell arrest were confirmed by the cell cycle report. At sub-G0/G1 phase, 2.10 ± 1.1 %; G0/G1 phase, 34.13 ± 1.9 %; S phase, 34.55 ± 0.98 %; and G2/M phase, 26.24 ± 1.7 % of cell arrest were observed. The results demonstrated the effectiveness of the proposed design for the development of corn starch-coated QT-NLs and their activity in breast cancer cell lines.
Collapse
Affiliation(s)
- E Bhargav
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, Andhra Pradesh, India
| | - Nawaz Mohammed
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, Andhra Pradesh, India
| | - Udit Narayan Singh
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, Andhra Pradesh, India
| | - P Ramalingam
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar, India
| | - Ranadheer Reddy Challa
- Formulation and Development, Quotient Sciences, 3080 McCann Farm Dr, Garnet Valley, PA, USA
| | | | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh-11451, Saudi Arabia
| | - Prasanth Dsnbk
- School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Jadcherla, Hyderabad, India
| | - Praveen Kumar Pasala
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, Andhra Pradesh, India
| | - Mithun Rudrapal
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical Sciences, Vignan's Foundation for Science, Technology & Research, Guntur, Andhra Pradesh, India
| |
Collapse
|
34
|
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
|
35
|
Gu Q, Zhu L. Heating Induced Nanoparticle Migration and Enhanced Delivery in Tumor Treatment Using Nanotechnology. Bioengineering (Basel) 2024; 11:900. [PMID: 39329642 PMCID: PMC11428587 DOI: 10.3390/bioengineering11090900] [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: 07/23/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Nanoparticles have been developed as imaging contrast agents, heat absorbers to confine energy into targeted tumors, and drug carriers in advanced cancer treatment. It is crucial to achieve a minimal concentration of drug-carrying nanostructures or to induce an optimized nanoparticle distribution in tumors. This review is focused on understanding how local or whole-body heating alters transport properties in tumors, therefore leading to enhanced nanoparticle delivery or optimized nanoparticle distributions in tumors. First, an overview of cancer treatment and the development of nanotechnology in cancer therapy is introduced. Second, the importance of particle distribution in one of the hyperthermia approaches using nanoparticles in damaging tumors is discussed. How intensive heating during nanoparticle hyperthermia alters interstitial space structure to induce nanoparticle migration in tumors is evaluated. The next section reviews major obstacles in the systemic delivery of therapeutic agents to targeted tumors due to unique features of tumor microenvironments. Experimental observations on how mild local or whole-body heating boosts systemic nanoparticle delivery to tumors are presented, and possible physiological mechanisms are explored. The end of this review provides the current challenges facing clinicians and researchers in designing effective and safe heating strategies to maximize the delivery of therapeutic agents to tumors.
Collapse
Affiliation(s)
- Qimei Gu
- Mechanical Engineering Department, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - Liang Zhu
- Mechanical Engineering Department, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| |
Collapse
|
36
|
Mohammed H, Karhib MM, Al-Fahad KSJ, Atef AM, Eskandrani A, Darwish AAE, Sary AA, Elwakil BH, Bakr BA, Eldrieny AM. Newly synthesized chitosan nanoparticles loaded with caffeine/moringa leaf extracts Halt Her2, BRCA1, and BRCA2 expressions. Sci Rep 2024; 14:18118. [PMID: 39103402 PMCID: PMC11300450 DOI: 10.1038/s41598-024-67599-1] [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: 09/28/2023] [Accepted: 07/12/2024] [Indexed: 08/07/2024] Open
Abstract
Breast cancer is among the highest morbidity and mortality rates in women around the world. In the present investigation we aimed to synthesis novel nanosystem combining two naturally important anticancer agents with different mechanism of action namely Moringa oleifera and caffeine. Firstly, chemical analysis of Moringa oleifera extract and caffeine was done by gas chromatography-mass spectroscopy (GC-MS) in order to assess the main chemical compounds present and correlate between them and the possible anticancer effect. The novel nanosystem was characterized through dynamic light scattering techniques which revealed the stability and homogeneity of the prepared M. oleifera leaves extract/Caffeine loaded chitosan nanoparticles, while FTIR and transmission electron microscope (TEM) proved the shape and the successful incorporation of M. oleifera leaves extract/Caffeine onto the nanochitosan carrier. Our initial step was to assess the anticancer effect in vitro in cancer cell line MCF-7 which proved the significant enhanced effect of M. oleifera leaves extract/Caffeine nanosystem compared to M. oleifera leaves extract or caffeine loaded nanoparticles. Further studies were conducted in vivo namely tumor biomarkers, tumor volume, bioluminescence imaging, molecular and histopathological investigations. The present study proved the potent anticancer effect of the synthesized M. oleifera leaves extract/Caffeine loaded chitosan nanoparticles. Mo/Caf/CsNPs exhibited a large number of apoptotic cells within the tumor mass while the adipose tissue regeneration was higher compared to the positive control. The prepared nanoparticles downregulated the expression of Her2, BRCA1 and BRCA2 while mTOR expression was upregulated. The aforementioned data demonstrated the successful synergistic impact of Moringa and caffeine in decreasing the carcinoma grade.
Collapse
Affiliation(s)
- Hanaa Mohammed
- Human Anatomy and Embryology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Mustafa M Karhib
- Department of Medical Laboratory Techniques, College of Health and Medical Technologies, Al-Mustaqbal University, Hillah, Babylon, 51001, Iraq
| | | | - Atef Mohamed Atef
- Faculty of Medical Applied Science, Irbid National University, Irbid, Jordan
| | - Areej Eskandrani
- College of Science, Taibah University, 30002, Madinah, Kingdom of Saudi Arabia
| | - Amira Abd-Elfattah Darwish
- Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, 21526, Egypt
| | - Ahmed Abdallah Sary
- Faculty of Physical Therapy, Pharos University in Alexandria, Alexandria, 21526, Egypt
| | - Bassma H Elwakil
- Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, 21526, Egypt.
| | - Basant A Bakr
- Faculty of Science, Alexandria University, Alexandria, 21321, Egypt
| | - Ahmed M Eldrieny
- Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, 21526, Egypt
| |
Collapse
|
37
|
Zhu M, Wu Y, Zhu T, Chen J, Chen Z, Ding H, Tan S, He J, Zeng Q, Huang X. Multifunctional Bispecific Nanovesicles Targeting SLAMF7 Trigger Potent Antitumor Immunity. Cancer Immunol Res 2024; 12:1007-1021. [PMID: 38819238 DOI: 10.1158/2326-6066.cir-23-1102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/03/2024] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
The effectiveness of immune checkpoint inhibitor (ICI) therapy is hindered by the ineffective infiltration and functioning of cytotoxic T cells and the immunosuppressive tumor microenvironment (TME). Signaling lymphocytic activation molecule family member 7 (SLAMF7) is a pivotal co-stimulatory receptor thought to simultaneously trigger NK-cell, T-cell, and macrophage antitumor cytotoxicity. However, the potential of this collaborative immune stimulation in antitumor immunity for solid tumors is underexplored due to the exclusive expression of SLAMF7 by hematopoietic cells. Here, we report the development and characterization of multifunctional bispecific nanovesicles (NVs) targeting SLAMF7 and glypican-3-a hepatocellular carcinoma (HCC)-specific tumor antigen. We found that by effectively "decorating" the surfaces of solid tumors with SLAMF7, these NVs directly induced potent and specific antitumor immunity and remodeled the immunosuppressive TME, sensitizing the tumors to programmed cell death protein 1 (PD1) blockade. Our findings highlight the potential of SLAMF7-targeted multifunctional bispecific NVs as an anticancer strategy with implications for designing next-generation targeted cancer therapies.
Collapse
Affiliation(s)
- Manman Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Tianchuan Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jian Chen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhenxing Chen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Kingcell Regenerative Medicine (Guangdong) Co., Zhuhai, China
| | - Hanxi Ding
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Siyi Tan
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jianzhong He
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qi Zeng
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
38
|
Zhang P, Cheng M, Levi-Kalisman Y, Raviv U, Xu Y, Han J, Dou H. Macromolecular Nano-Assemblies for Enhancing the Effect of Oxygen-Dependent Photodynamic Therapy Against Hypoxic Tumors. Chemistry 2024; 30:e202401700. [PMID: 38797874 DOI: 10.1002/chem.202401700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
In oxygen (O2)-dependent photodynamic therapy (PDT), photosensitizers absorb light energy, which is then transferred to ambient O2 and subsequently generates cytotoxic singlet oxygen (1O2). Therefore, the availability of O2 and the utilization efficiency of generated 1O2 are two significant factors that influence the effectiveness of PDT. However, tumor microenvironments (TMEs) characterized by hypoxia and limited utilization efficiency of 1O2 resulting from its short half-life and short diffusion distance significantly restrict the applicability of PDT for hypoxic tumors. To address these challenges, numerous macromolecular nano-assemblies (MNAs) have been designed to relieve hypoxia, utilize hypoxia or enhance the utilization efficiency of 1O2. Herein, we provide a comprehensive review on recent advancements achieved with MNAs in enhancing the effectiveness of O2-dependent PDT against hypoxic tumors.
Collapse
Affiliation(s)
- Peipei Zhang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, China
| | - Meng Cheng
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, China
| | - Yael Levi-Kalisman
- Institute of Life Sciences and the Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond Safra Campus, 9190401, Givat Ram, Jerusalem, Israel
| | - Uri Raviv
- Institute of Chemistry and the Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Edmond Safra Campus, 9190401, Givat Ram, Jerusalem, Israel
| | - Yichun Xu
- Shanghai Biochip Co. Ltd. and National Engineering Center for Biochip at Shanghai, 151 Libing Road, 201203, Shanghai, China
| | - Junsong Han
- Shanghai Biochip Co. Ltd. and National Engineering Center for Biochip at Shanghai, 151 Libing Road, 201203, Shanghai, China
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, China
| |
Collapse
|
39
|
Li M, Guo X, Verma A, Rudkouskaya A, McKenna AM, Intes X, Wang G, Barroso M. Contrast-enhanced photon-counting micro-CT of tumor xenograft models. Phys Med Biol 2024; 69:155011. [PMID: 38670143 PMCID: PMC11258216 DOI: 10.1088/1361-6560/ad4447] [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: 01/12/2024] [Revised: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 04/28/2024]
Abstract
Objective. Photon-counting micro-computed tomography (micro-CT) is a major advance in small animal preclinical imaging. Small molecule- and nanoparticle-based contrast agents have been widely used to enable the differentiation of liver tumors from surrounding tissues using photon-counting micro-CT. However, there is a notable gap in the application of these market-available agents to the imaging of breast and ovarian tumors using photon-counting micro-CT. Herein, we have used photon-counting micro-CT to determine the effectiveness of these contrast agents in differentiating ovarian and breast tumor xenografts in live, intact mice.Approach. Nude mice carrying different types of breast and ovarian tumor xenografts (AU565, MDA-MB-231 and SKOV-3 human cancer cells) were injected with ISOVUE-370 (a small molecule-based agent) or Exitron Nano 12000 (a nanoparticle-based agent) and subjected to photon-counting micro-CT. To improve tumor visualization using photon-counting micro-CT, we developed a novel color visualization method, which changes color tones to highlight contrast media distribution, offering a robust alternative to traditional material decomposition methods with less computational demand.Main results. Ourin vivoexperiments confirm the effectiveness of this color visualization approach, showing distinct enhancement characteristics for each contrast agent. Qualitative and quantitative analyses suggest that Exitron Nano 12000 provides superior vasculature enhancement and better quantitative consistency across scans, while ISOVUE-370 delivers a more comprehensive tumor enhancement but with significant variance between scans due to its short blood half-time. Further, a paired t-test on mean and standard deviation values within tumor volumes showed significant differences between the AU565 and SKOV-3 tumor models with the nanoparticle-based contrast agent (p-values < 0.02), attributable to their distinct vascularity, as confirmed by immunohistochemical analysis.Significance. These findings underscore the utility of photon-counting micro-CT in non-invasively assessing the morphology and anatomy of different tumor xenografts, which is crucial for tumor characterization and longitudinal monitoring of tumor progression and response to treatments.
Collapse
Affiliation(s)
- Mengzhou Li
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Xiaodong Guo
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Amit Verma
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| | - Antigone M McKenna
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Xavier Intes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Ge Wang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| |
Collapse
|
40
|
Liang Y, Fan T, Bai M, Cui N, Li W, Wang J, Guan Y. Chikusetsu Saponin IVa liposomes modified with a retro-enantio peptide penetrating the blood-brain barrier to suppress pyroptosis in acute ischemic stroke rats. J Nanobiotechnology 2024; 22:393. [PMID: 38965602 PMCID: PMC11223377 DOI: 10.1186/s12951-024-02641-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND The therapeutic strategies for acute ischemic stroke were faced with substantial constraints, emphasizing the necessity to safeguard neuronal cells during cerebral ischemia to reduce neurological impairments and enhance recovery outcomes. Despite its potential as a neuroprotective agent in stroke treatment, Chikusetsu saponin IVa encounters numerous challenges in clinical application. RESULT Brain-targeted liposomes modified with THRre peptides showed substantial uptake by bEnd. 3 and PC-12 cells and demonstrated the ability to cross an in vitro blood-brain barrier model, subsequently accumulating in PC-12 cells. In vivo, they could significantly accumulate in rat brain. Treatment with C-IVa-LPs-THRre notably reduced the expression of proteins in the P2RX7/NLRP3/Caspase-1 pathway and inflammatory factors. This was evidenced by decreased cerebral infarct size and improved neurological function in MCAO rats. CONCLUSION The findings indicate that C-IVa-LPs-THRre could serve as a promising strategy for targeting cerebral ischemia. This approach enhances drug concentration in the brain, mitigates pyroptosis, and improves the neuroinflammatory response associated with stroke.
Collapse
Affiliation(s)
- Yitong Liang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Tingting Fan
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Min Bai
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Na Cui
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Wangting Li
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China.
| | - Yue Guan
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China.
| |
Collapse
|
41
|
Aloss K, Hamar P. Augmentation of the EPR effect by mild hyperthermia to improve nanoparticle delivery to the tumor. Biochim Biophys Acta Rev Cancer 2024; 1879:189109. [PMID: 38750699 DOI: 10.1016/j.bbcan.2024.189109] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
The clinical translation of the nanoparticle (NP)-based anticancer therapies is still unsatisfactory due to the heterogeneity of the enhanced permeability and retention (EPR) effect. Despite the promising preclinical outcome of the pharmacological EPR enhancers, their systemic toxicity can limit their clinical application. Hyperthermia (HT) presents an efficient tool to augment the EPR by improving tumor blood flow (TBF) and vascular permeability, lowering interstitial fluid pressure (IFP), and disrupting the structure of the extracellular matrix (ECM). Furthermore, the HT-triggered intravascular release approach can overcome the EPR effect. In contrast to pharmacological approaches, HT is safe and can be focused to cancer tissues. Moreover, HT conveys direct anti-cancer effects, which improve the efficacy of the anti-cancer agents encapsulated in NPs. However, the clinical application of HT is challenging due to the heterogeneous distribution of temperature within the tumor, the length of the treatment and the complexity of monitoring.
Collapse
Affiliation(s)
- Kenan Aloss
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary
| | - Péter Hamar
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary.
| |
Collapse
|
42
|
Canlas KKV, Park H. Applications of Biomolecular Nanostructures for Anti-Angiogenic Theranostics. Int J Nanomedicine 2024; 19:6485-6497. [PMID: 38946886 PMCID: PMC11214753 DOI: 10.2147/ijn.s459928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/10/2024] [Indexed: 07/02/2024] Open
Abstract
Angiogenesis is a physiological process of forming new blood vessels that has pathological importance in seemingly unrelated illnesses like cancer, diabetes, and various inflammatory diseases. Treatment targeting angiogenesis has shown promise for these types of diseases, but current anti-angiogenic agents have critical limitations in delivery and side-effects. This necessitates exploration of alternative approaches like biomolecule-based drugs. Proteins, lipids, and oligonucleotides have recently become popular in biomedicine, specifically as biocompatible components of therapeutic drugs. Their excellent bioavailability and potential bioactive and immunogenic properties make them prime candidates for drug discovery or drug delivery systems. Lipid-based liposomes have become standard vehicles for targeted nanoparticle (NP) delivery, while protein and nucleotide NPs show promise for environment-sensitive delivery as smart NPs. Their therapeutic applications have initially been hampered by short circulation times and difficulty of fabrication but recent developments in nanofabrication and NP engineering have found ways to circumvent these disadvantages, vastly improving the practicality of biomolecular NPs. In this review, we are going to briefly discuss how biomolecule-based NPs have improved anti-angiogenesis-based therapy.
Collapse
Affiliation(s)
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Korea
| |
Collapse
|
43
|
Oopkaew L, Injongkol Y, Rimsueb N, Mahalapbutr P, Choowongkomon K, Hadsadee S, Rojanathanes R, Rungrotmongkol T. Targeted Therapy with Cisplatin-Loaded Calcium Citrate Nanoparticles Conjugated with Epidermal Growth Factor for Lung Cancer Treatment. ACS OMEGA 2024; 9:25668-25677. [PMID: 38911765 PMCID: PMC11191089 DOI: 10.1021/acsomega.3c08969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 05/19/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide with high incidence rates for new cases. Conventional cisplatin (CDDP) therapy has limitations due to severe side effects from nonspecific targeting. To address this challenge, nanomedicine offers targeted therapies. In this study, cisplatin-loaded calcium citrate nanoparticles conjugated with epidermal growth factor (CaCit@CDDP-EGF NPs) were synthesized. The resulting nanodrug had a size below 350 nm with a cation charge. Based on density functional theory (DFT), the CaCit@CDDP NP model containing two citrates substituted on two chlorides exhibited a favorable binding energy of -5.42 eV, and the calculated spectrum at 261 nm closely matched the experimental data. CaCit@CDDP-EGF NPs showed higher inhibition rates against EGFR-expressed and mutant carcinoma cells compared to those of cisplatin while displaying lower cytotoxicity to lung fibroblast cells. Integrating in vitro experiments with in silico studies, these nanoparticles hold promise as a novel nanomedicine for targeted therapy in clinical applications.
Collapse
Affiliation(s)
- Lipika Oopkaew
- Center
of Excellence in Biocatalyst and Sustainable Biotechnology, Department
of Biochemistry, Faculty of Science, Chulalongkorn
University, Bangkok 10330, Thailand
| | - Yuwanda Injongkol
- Center
of Excellence in Biocatalyst and Sustainable Biotechnology, Department
of Biochemistry, Faculty of Science, Chulalongkorn
University, Bangkok 10330, Thailand
| | - Natchanon Rimsueb
- National
Nanotechnology Center NANOTEC, National
Science and Technology Development Agency NSTDA, Pathum Thani 12120, Thailand
- Center
of Excellence in Nanomedicine, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panupong Mahalapbutr
- Department
of Biochemistry, Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Kiattawee Choowongkomon
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| | - Sarinya Hadsadee
- Center
of Excellence in Biocatalyst and Sustainable Biotechnology, Department
of Biochemistry, Faculty of Science, Chulalongkorn
University, Bangkok 10330, Thailand
| | - Rojrit Rojanathanes
- Center of
Excellence in Biocatalyst and Sustainable Biotechnology, Department
of Chemistry, Faculty of Science, Chulalongkorn
University Bangkok, 10330, Thailand
| | - Thanyada Rungrotmongkol
- Center
of Excellence in Biocatalyst and Sustainable Biotechnology, Department
of Biochemistry, Faculty of Science, Chulalongkorn
University, Bangkok 10330, Thailand
- Program
in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
44
|
Slayden O, Luo F, Park Y, Moses AS, Demessie AA, Singh P, Korzun T, Taratula O, Taratula O. Targeted nanoparticles for imaging and therapy of endometriosis†. Biol Reprod 2024; 110:1191-1200. [PMID: 38738758 PMCID: PMC11180615 DOI: 10.1093/biolre/ioae073] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024] Open
Abstract
In this brief review, we discuss our efforts to validate nanoplatforms for imaging and treatment of endometriosis. We specifically highlight our use of nonhuman primates and primate tissues in this effort. Endometriosis is a painful disorder of women and nonhuman primates where endometrium-like tissue exists outside of the uterus. There are no reliable, specific, and noninvasive diagnostic tests for endometriosis. Laparoscopic imaging remains the gold standard for identifying small endometriotic lesions in both women and monkeys. Visualizing and surgically removing microscopic lesions remains a clinical challenge. To address this challenge, we have created nanoparticle reagents that, when administered intravenously, enter endometriotic lesions both passively and by targeting endometriotic cells. The particles can carry payloads, including near-infrared fluorescent dyes and magnetic nanoparticles. These agents can be used for imaging and thermal ablation of diseased tissues. We evaluated this approach on macaque endometriotic cells, human and macaque endometrium engrafted into immunodeficient mice, in endometrium subcutaneously autografted in macaques, and in rhesus monkeys with spontaneous endometriosis. Employing these models, we report that nanoplatform-based reagents can improve imaging and provide thermal ablation of endometriotic tissues.
Collapse
Affiliation(s)
- Ov Slayden
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Fangzhou Luo
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Youngrong Park
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Ananiya A Demessie
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Prem Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
- School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Olena Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, USA
| |
Collapse
|
45
|
ter Linden E, Abels ER, van Solinge TS, Neefjes J, Broekman MLD. Overcoming Barriers in Glioblastoma-Advances in Drug Delivery Strategies. Cells 2024; 13:998. [PMID: 38920629 PMCID: PMC11201826 DOI: 10.3390/cells13120998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
The world of cancer treatment is evolving rapidly and has improved the prospects of many cancer patients. Yet, there are still many cancers where treatment prospects have not (or hardly) improved. Glioblastoma is the most common malignant primary brain tumor, and even though it is sensitive to many chemotherapeutics when tested under laboratory conditions, its clinical prospects are still very poor. The blood-brain barrier (BBB) is considered at least partly responsible for the high failure rate of many promising treatment strategies. We describe the workings of the BBB during healthy conditions and within the glioblastoma environment. How the BBB acts as a barrier for therapeutic options is described as well as various approaches developed and tested for passing or opening the BBB, with the ultimate aim to allow access to brain tumors and improve patient perspectives.
Collapse
Affiliation(s)
- Esther ter Linden
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.t.L.); (E.R.A.)
| | - Erik R. Abels
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.t.L.); (E.R.A.)
| | - Thomas S. van Solinge
- Department of Neurosurgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Jacques Neefjes
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.t.L.); (E.R.A.)
| | - Marike L. D. Broekman
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (E.t.L.); (E.R.A.)
- Department of Neurosurgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Department of Neurosurgery, Haaglanden Medical Center, 2512 VA The Hague, The Netherlands
| |
Collapse
|
46
|
Paramshetti S, Angolkar M, Talath S, Osmani RAM, Spandana A, Al Fatease A, Hani U, Ramesh KVRNS, Singh E. Unravelling the in vivo dynamics of liposomes: Insights into biodistribution and cellular membrane interactions. Life Sci 2024; 346:122616. [PMID: 38599316 DOI: 10.1016/j.lfs.2024.122616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
Liposomes, as a colloidal drug delivery system dating back to the 1960s, remain a focal point of extensive research and stand as a highly efficient drug delivery method. The amalgamation of technological and biological advancements has propelled their evolution, elevating them to their current status. The key attributes of biodegradability and biocompatibility have been instrumental in driving substantial progress in liposome development. Demonstrating a remarkable ability to surmount barriers in drug absorption, enhance stability, and achieve targeted distribution within the body, liposomes have become pivotal in pharmaceutical research. In this comprehensive review, we delve into the intricate details of liposomal drug delivery systems, focusing specifically on their pharmacokinetics and cell membrane interactions via fusion, lipid exchange, endocytosis etc. Emphasizing the nuanced impact of various liposomal characteristics, we explore factors such as lipid composition, particle size, surface modifications, charge, dosage, and administration routes. By dissecting the multifaceted interactions between liposomes and biological barriers, including the reticuloendothelial system (RES), opsonization, enhanced permeability and retention (EPR) effect, ATP-binding cassette (ABC) phenomenon, and Complement Activation-Related Pseudoallergy (CARPA) effect, we provide a deeper understanding of liposomal behaviour in vivo. Furthermore, this review addresses the intricate challenges associated with translating liposomal technology into practical applications, offering insights into overcoming these hurdles. Additionally, we provide a comprehensive analysis of the clinical adoption and patent landscape of liposomes across diverse biomedical domains, shedding light on their potential implications for future research and therapeutic developments.
Collapse
Affiliation(s)
- Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - K V R N S Ramesh
- Department of Pharmaceutics, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Ekta Singh
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
47
|
Kim D, Javius-Jones K, Mamidi N, Hong S. Dendritic nanoparticles for immune modulation: a potential next-generation nanocarrier for cancer immunotherapy. NANOSCALE 2024; 16:10208-10220. [PMID: 38727407 DOI: 10.1039/d4nr00635f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Immune activation, whether occurring from direct immune checkpoint blockade or indirectly as a result of chemotherapy, is an approach that has drastically impacted the way we treat cancer. Utilizing patients' own immune systems for anti-tumor efficacy has been translated to robust immunotherapies; however, clinically significant successes have been achieved in only a subset of patient populations. Dendrimers and dendritic polymers have recently emerged as a potential nanocarrier platform that significantly improves the therapeutic efficacy of current and next-generation cancer immunotherapies. In this paper, we highlight the recent progress in developing dendritic polymer-based therapeutics with immune-modulating properties. Specifically, dendrimers, dendrimer hybrids, and dendronized copolymers have demonstrated promising results and are currently in pre-clinical development. Despite their early stage of development, these nanocarriers hold immense potential to make profound impact on cancer immunotherapy and combination therapy. This overview provides insights into the potential impact of dendrimers and dendron-based polymers, offering a preview of their potential utilities for various aspects of cancer treatment.
Collapse
Affiliation(s)
- DaWon Kim
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
| | - Narsimha Mamidi
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, USA
- Lachman Institute for Drug Development, University of Wisconsin-Madison, Madison, WI, USA
- Yonsei Frontier Lab, Yonsei University, Seoul, Korea
| |
Collapse
|
48
|
Petrovic S, Bita B, Barbinta-Patrascu ME. Nanoformulations in Pharmaceutical and Biomedical Applications: Green Perspectives. Int J Mol Sci 2024; 25:5842. [PMID: 38892030 PMCID: PMC11172476 DOI: 10.3390/ijms25115842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
This study provides a brief discussion of the major nanopharmaceuticals formulations as well as the impact of nanotechnology on the future of pharmaceuticals. Effective and eco-friendly strategies of biofabrication are also highlighted. Modern approaches to designing pharmaceutical nanoformulations (e.g., 3D printing, Phyto-Nanotechnology, Biomimetics/Bioinspiration, etc.) are outlined. This paper discusses the need to use natural resources for the "green" design of new nanoformulations with therapeutic efficiency. Nanopharmaceuticals research is still in its early stages, and the preparation of nanomaterials must be carefully considered. Therefore, safety and long-term effects of pharmaceutical nanoformulations must not be overlooked. The testing of nanopharmaceuticals represents an essential point in their further applications. Vegetal scaffolds obtained by decellularizing plant leaves represent a valuable, bioinspired model for nanopharmaceutical testing that avoids using animals. Nanoformulations are critical in various fields, especially in pharmacy, medicine, agriculture, and material science, due to their unique properties and advantages over conventional formulations that allows improved solubility, bioavailability, targeted drug delivery, controlled release, and reduced toxicity. Nanopharmaceuticals have transitioned from experimental stages to being a vital component of clinical practice, significantly improving outcomes in medical fields for cancer treatment, infectious diseases, neurological disorders, personalized medicine, and advanced diagnostics. Here are the key points highlighting their importance. The significant challenges, opportunities, and future directions are mentioned in the final section.
Collapse
Affiliation(s)
- Sanja Petrovic
- Department of Chemical Technologies, Faculty of Technology, University of Nis, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia;
| | - Bogdan Bita
- Department of Electricity, Solid-State Physics and Biophysics, Faculty of Physics, University of Bucharest, 405 Atomistilor Street, P.O. Box MG-11, 077125 Magurele, Romania;
| | - Marcela-Elisabeta Barbinta-Patrascu
- Department of Electricity, Solid-State Physics and Biophysics, Faculty of Physics, University of Bucharest, 405 Atomistilor Street, P.O. Box MG-11, 077125 Magurele, Romania;
| |
Collapse
|
49
|
Dacos M, Immordino B, Diroff E, Sicard G, Kosta A, Rodallec A, Giacometti S, Ciccolini J, Fanciullino R. Pegylated liposome encapsulating docetaxel using microfluidic mixing technique: Process optimization and results in breast cancer models. Int J Pharm 2024; 656:124091. [PMID: 38588758 DOI: 10.1016/j.ijpharm.2024.124091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024]
Abstract
The development of nanoparticles could help to improve the efficacy/toxicity balance of drugs. This project aimed to develop liposomes and immunoliposomes using microfluidic mixing technology.Various formulation tests were carried out to obtain liposomes that met the established specifications. The liposomes were then characterized in terms of size, polydispersity index (PDI), docetaxel encapsulation rate and lamellarity. Antiproliferative activity was tested in human breast cancer models ranging from near-negative (MDA-MB-231), positive (MDA-MB-453) to HER2 positive. Pharmacokinetic studies were performed in C57BL/6 mice.Numerous batches of liposomes were synthesised using identical molar ratios and by varying the microfluidic parameters TFR, FRR and buffer. All synthesized liposomes have a size < 200 nm, but only Lipo-1, Lipo-6, Lipo-7, Lipo-8 have a PDI < 0.2, which meets our initial requirements. The size of the liposomes was correlated with the total FRR, for a 1:1 FRR the size is 122.2 ± 12.3 nm, whereas for a 1:3 FRR the size obtained is 163.4 ± 34.0 nm (p = 0.019. Three batches of liposomes were obtained with high docetaxel encapsulation rates > 80 %. Furthermore, in vitro studies on breast cancer cell lines demonstrated the efficacy of liposomes obtained by microfluidic mixing technique. These liposomes also showed improved pharmacokinetics compared to free docetaxel, with a longer half-life and higher AUC (3-fold and 3.5-fold increase for the immunoliposome, respectively).This suggests that switching to the microfluidic process will produce batches of liposomes with the same characteristics in terms of in vitro properties and efficacy, as well as the ability to release the encapsulated drug over time in vivo. This time-efficiency of the microfluidic technique is critical, especially in the early stages of development.
Collapse
Affiliation(s)
- Mathilde Dacos
- COMPO, SMARTc. CRCM: UMR Inserm 1068, CNRS UMR 7258, AMU U105, IPC, Marseille, France; Assitance Publique des Hôpitaux de Marseille, Marseille, France.
| | - Benoît Immordino
- Fondazione Pisana per La Scienza, 56017 San Giuliano, Pisa, Italy
| | - Erwan Diroff
- COMPO, SMARTc. CRCM: UMR Inserm 1068, CNRS UMR 7258, AMU U105, IPC, Marseille, France
| | - Guillaume Sicard
- COMPO, SMARTc. CRCM: UMR Inserm 1068, CNRS UMR 7258, AMU U105, IPC, Marseille, France; Assitance Publique des Hôpitaux de Marseille, Marseille, France
| | - Artemis Kosta
- Microscopy Core Facility, Institut de Microbiologie de la Méditerranée (FR3479), CNRS, Aix-Marseille Université, Marseille, France
| | - Anne Rodallec
- COMPO, SMARTc. CRCM: UMR Inserm 1068, CNRS UMR 7258, AMU U105, IPC, Marseille, France
| | - Sarah Giacometti
- COMPO, SMARTc. CRCM: UMR Inserm 1068, CNRS UMR 7258, AMU U105, IPC, Marseille, France
| | - Joseph Ciccolini
- COMPO, SMARTc. CRCM: UMR Inserm 1068, CNRS UMR 7258, AMU U105, IPC, Marseille, France; Assitance Publique des Hôpitaux de Marseille, Marseille, France
| | - Raphaëlle Fanciullino
- COMPO, SMARTc. CRCM: UMR Inserm 1068, CNRS UMR 7258, AMU U105, IPC, Marseille, France; Assitance Publique des Hôpitaux de Marseille, Marseille, France
| |
Collapse
|
50
|
Xu D, Li Y, Yin S, Huang F. Strategies to address key challenges of metallacycle/metallacage-based supramolecular coordination complexes in biomedical applications. Chem Soc Rev 2024; 53:3167-3204. [PMID: 38385584 DOI: 10.1039/d3cs00926b] [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: 02/23/2024]
Abstract
Owing to their capacity for dynamically linking two or more functional molecules, supramolecular coordination complexes (SCCs), exemplified by two-dimensional (2D) metallacycles and three-dimensional (3D) metallacages, have gained increasing significance in biomedical applications. However, their inherent hydrophobicity and self-assembly driven by heavy metal ions present common challenges in their applications. These challenges can be overcome by enhancing the aqueous solubility and in vivo circulation stability of SCCs, alongside minimizing their side effects during treatment. Addressing these challenges is crucial for advancing the fundamental research of SCCs and their subsequent clinical translation. In this review, drawing on extensive contemporary research, we offer a thorough and systematic analysis of the strategies employed by SCCs to surmount these prevalent yet pivotal obstacles. Additionally, we explore further potential challenges and prospects for the broader application of SCCs in the biomedical field.
Collapse
Affiliation(s)
- Dongdong Xu
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China.
| | - Yang Li
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China.
| | - Shouchun Yin
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China.
| | - Feihe Huang
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou 310058, P. R. China.
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, P. R. China
| |
Collapse
|