1
|
Panda P, Mohanty S, Gouda SR, Mohapatra R. Advances in nanomedicine for retinal drug delivery: overcoming barriers and enhancing therapeutic outcomes. J Drug Target 2025; 33:587-611. [PMID: 39694681 DOI: 10.1080/1061186x.2024.2443144] [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: 10/22/2024] [Revised: 11/16/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
Nanomedicine offers a promising avenue for improving retinal drug delivery, effectively addressing challenges associated with ocular diseases like age-related macular degeneration and diabetic retinopathy. Nanoparticles, with their submicron size and customisable surface properties, enable enhanced permeability and retention within retinal tissues, supporting sustained drug release and minimising systemic side effects. Nanostructured scaffolds further provide a supportive environment for retinal cell growth and tissue regeneration, crucial for treating degenerative conditions. Additionally, advanced nanodevices facilitate real-time monitoring and controlled drug release, marking significant progress in retinal therapy. This study reviews recent advancements in nanomedicine for retinal drug delivery, critically analysing design innovations, therapeutic benefits, and limitations of these systems. By advancing nanotechnology integration in ocular therapies, this field holds strong potential for overcoming current barriers, ultimately improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Pratikeswar Panda
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Shreyashree Mohanty
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Sangita Ranee Gouda
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Rajaram Mohapatra
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| |
Collapse
|
2
|
Al-Ani T, Lupton K. Ultrasound-guided intra-articular injection therapy. BJA Educ 2025; 25:199-205. [PMID: 40256652 PMCID: PMC12009082 DOI: 10.1016/j.bjae.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2025] [Indexed: 04/22/2025] Open
Affiliation(s)
- T. Al-Ani
- Glasgow Royal Infirmary, Glasgow, UK
| | - K. Lupton
- Glasgow Royal Infirmary, Glasgow, UK
| |
Collapse
|
3
|
Tihăuan BM, Onisei T, Slootweg W, Gună D, Iliescu C, Chifiriuc MC. Cannabidiol-A friend or a foe? Eur J Pharm Sci 2025; 208:107036. [PMID: 39929375 DOI: 10.1016/j.ejps.2025.107036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 03/23/2025]
Abstract
Cannabidiol (CBD), one of the main actives from Cannabis sativa has been perpetually explored lately for its therapeutic effects. Its main attributes, such as anti-inflammatory and antioxidant effects, snowball into pain management, epilepsy and seizure alleviation, anxiety relief, as well as numerous other implications through the entire metabolism. However, conventional administration routes challenge its therapeutic potential, with reported poor water solubility, hepatic degradation, gastric instability and erratic bioavailability observed in oral administration. As a result, the transdermal delivery systems have emerged as a promising alternative to oral or inhaled routes, offering improved bioavailability and targeted effects. The medical use of CBD throughout Europe, UK, USA or Australia is extensive and usually represented by pharmaceutical preparations recommended after conventional treatment routs fail. The non-medical use is limited by each country's own legislation, a wider range of products being available, but the irregular regulatory landscape coupled with the growing market of cannabinoid-infused products, emphasizes the need for standardized formulations and further clinical research. The present work critically examines the transdermal administration of cannabidiol, explores the skin's potential as a route and the strategies involved in using it for systemic targeting. We highlighted key challenges and provided insights into CBD`s variable bioavailability based on different administration routes and methods, thus compiling a literature-based absorption, distribution, metabolism, and excretion (ADME) study. We also explore the role of the endocannabinoid system, its function in various medical conditions, and the therapeutic effects associated with CBD, particularly in light of the varying legislation across countries. While the breadth of potential benefits is compelling, it is essential to emphasize the ongoing nature of CBD research as individual responses to it can vary significantly.
Collapse
Affiliation(s)
- Bianca-Maria Tihăuan
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 50567 Bucharest, Romania; National Institute for Research and Development in Food Bioresources, Dinu Vintilă Street, No.6, 021102 Bucharest, Romania; eBio-hub Research Centre, National University for Science and Technology Politehnica Bucharest, Bucharest, Romania; Academy of Romanian Scientists, Bucharest, Romania
| | - Tatiana Onisei
- National Institute for Research and Development in Food Bioresources, Dinu Vintilă Street, No.6, 021102 Bucharest, Romania
| | - Walter Slootweg
- QB3 Research & Development, Spaarndammerstraaat 4d, 1013SV Amsterdam, Netherlands
| | - Daniel Gună
- S.C. Absolute Essential Oils Ltd. (AEO), Adunații Copăceni Village, Giurgiu County, 38 Troitei Street, 087005, Romania
| | - Ciprian Iliescu
- eBio-hub Research Centre, National University for Science and Technology Politehnica Bucharest, Bucharest, Romania; Academy of Romanian Scientists, Bucharest, Romania; National Institute for Microtechnologies, 126A Erou Iancu Nicolae Street, Voluntari 077190, Romania.
| | - Mariana-Carmen Chifiriuc
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 50567 Bucharest, Romania.
| |
Collapse
|
4
|
Franco PIR, do Carmo Neto JR, Braga YLL, de Lima Pedroso B, Martins JA, Rocha VL, Amaral AC, Martins DB, Ruiz KC, Pereira JX, Machado E Silva JR, Miguel MP. Melatonin-loaded lecithin and chitosan nanoparticles are cytotoxic to 4 T1 breast cancer cells and safe in a BALB/c mouse model. Int J Biol Macromol 2025:143401. [PMID: 40268017 DOI: 10.1016/j.ijbiomac.2025.143401] [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: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Melatonin is used as an adjuvant therapy in cancer treatment. However, its effectiveness is limited because of its low bioavailability. Polymeric nanoparticles (NPs) made of chitosan and lecithin have been developed to overcome this limitation and optimize localized drug delivery. These lecithin and chitosan-based NPs loaded with melatonin (NP-MEL) were evaluated for their cytotoxic potential in metastatic breast cancer cells and their safety profile in a murine model. Physicochemical characterization revealed efficient melatonin encapsulation (31 %), a positive zeta potential (48.6 mV), and controlled release at physiological pH. NP-MEL exhibited selective cytotoxicity in vitro, with a toxic concentration capable of killing 50 % of the cells (CC50) of 109.53 μg/mL for 4 T1 cancer cells and a significantly higher CC50 of 1460.59 μg/mL for normal VERO cells, resulting in a selectivity index of 13.33. In vivo experiments with BALB/c mice with tumor implantation treated with NP-MEL (2 mg/kg/day for 21 days) showed no significant changes in weight, clinical signs, or biochemical markers of liver and kidney function, except for changes in gamma-glutamyl transferase levels. Histopathological analyses confirmed the preservation of the liver and kidney architecture in the NP-MEL-treated group, in contrast to the moderate-to-severe kidney damage observed in animals treated with empty NPs. These findings highlight the low toxicity and therapeutic potential of NP-MEL as a controlled and targeted-release system for breast cancer treatment, indicating the need for further preclinical investigation.
Collapse
Affiliation(s)
- Pablo Igor Ribeiro Franco
- Instituto de Patologia Tropical e Saúde Pública, Laboratório de Histotécnica e Inovação, Centro Multiusuário de Pesquisa de Bioinsumos, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - José Rodrigues do Carmo Neto
- Instituto de Patologia Tropical e Saúde Pública, Laboratório de Histotécnica e Inovação, Centro Multiusuário de Pesquisa de Bioinsumos, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Yarlla Loyane Lira Braga
- Instituto de Patologia Tropical e Saúde Pública, Laboratório de Histotécnica e Inovação, Centro Multiusuário de Pesquisa de Bioinsumos, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Bárbara de Lima Pedroso
- Instituto de Patologia Tropical e Saúde Pública, Laboratório de Histotécnica e Inovação, Centro Multiusuário de Pesquisa de Bioinsumos, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Juliana Assis Martins
- Laboratório de Nano & Biotecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Viviane Lopes Rocha
- Laboratório de Nano & Biotecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Andre Correa Amaral
- Laboratório de Nano & Biotecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Danieli Brolo Martins
- Programa de Pós-Graduação em Ciência Animal, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Katalina Cifuentes Ruiz
- Programa de Pós-Graduação em Ciência Animal, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Jonathas Xavier Pereira
- Instituto de Patologia Tropical e Saúde Pública, Laboratório de Histotécnica e Inovação, Centro Multiusuário de Pesquisa de Bioinsumos, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Juliana Reis Machado E Silva
- Departamento de Patologia, Genética e Evolução, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Marina Pacheco Miguel
- Instituto de Patologia Tropical e Saúde Pública, Laboratório de Histotécnica e Inovação, Centro Multiusuário de Pesquisa de Bioinsumos, Universidade Federal de Goiás, Goiânia, GO, Brazil; Programa de Pós-Graduação em Ciência Animal, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
5
|
Jin H, Noh W, Kyung K, Yeo WS, Song YH, Heo YS, Kim DE. Aptamer- vs Fab-Conjugated Liposomes: A Comparative Study in Targeting Acute Myeloid Leukemia Cells. Bioconjug Chem 2025; 36:815-822. [PMID: 40148126 DOI: 10.1021/acs.bioconjchem.5c00065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy characterized by uncontrolled proliferation of abnormal myeloid cells with a generally poor prognosis despite advancements in chemotherapy and stem cell transplantation. To enhance therapeutic efficacy and minimize systemic toxicity, we designed liposomal nanoparticles functionalized with two distinct targeting ligands, a DNA aptamer or fragment-antigen-binding (Fab) antibody, targeting the surface marker transmembrane glycoprotein CD33 antigen (CD33) on AML cells. Aptamer- and Fab-conjugated liposomes (Apt-Lipm and Fab-Lipm, respectively) were prepared and tested for cellular uptake by CD33-positive AML cell lines. Comparative studies revealed that Fab-Lipm exhibited significantly superior binding affinity, targeting efficiency, and cellular uptake compared with Apt-Lipm. Furthermore, we demonstrated the intracellular distribution and endocytic pathways of Fab-Lipm during the cellular uptake. This comparative study of aptamer- and Fab-conjugated liposomes suggests that the Fab-conjugated liposomal system offers enhanced precision in targeting AML cells for the development of effective therapeutic strategies against hematologic malignancies.
Collapse
Affiliation(s)
- Hyesoo Jin
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Wooseong Noh
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Kangwuk Kyung
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Woon-Seok Yeo
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Ye Han Song
- Department of Chemistry, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Yong-Seok Heo
- Department of Chemistry, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Gwangjin-gu, Seoul05029, Republic of Korea
- Uniwon PharmGene Inc, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
6
|
Chang CY, Huang SH, Chen CY, Jian CB, Chang CC, Chang YY, Jung M, Lee HM, Cheng B. Monocyte-adhesive peptidyl liposomes for harnessing monocyte homing to tumor tissues. J Control Release 2025; 382:113672. [PMID: 40185332 DOI: 10.1016/j.jconrel.2025.113672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In current drug delivery strategies, the efficiency of most carriers still largely depends on their ability to passively infiltrate target tissues. To overcome this limitation, we developed monocyte-adhesive peptidyl liposomes, termed monocyte-mediated drug carriers (MMDCs). These carriers are designed to exploit the innate chemotactic properties of monocytes, which actively home to diseased tissues. MMDCs were shown to effectively hitchhike on circulating monocytes (THP-1 cells) under physiological flow conditions. Their targeting specificity was further demonstrated in a 3D microfluidic culture system consisting of human breast cancer spheroids (MDA-MB-231) embedded in a collagen matrix, overlaid with a human endothelial cell-derived barrier. MMDCs underwent trans-endothelial migration via monocyte hitchhiking and selectively recognized collagen matrices containing MDA-MB-231 cells, but not those embedded with non-cancerous cells. In vitro assays revealed that doxorubicin encapsulated in MMDCs was released into the extracellular environment following phagocytosis of the carriers by THP-1-derived macrophages. In a xenograft mouse model, MMDCs exhibited high tumor-targeting efficiency. By harnessing the homing capability of monocytes, MMDCs significantly improved drug biodistribution at the disease site, thereby enhancing the therapeutic efficacy of the encapsulated agents.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, Taichung, Taiwan
| | - Shih-Hsun Huang
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, Taichung, Taiwan
| | | | - Cheng-Bang Jian
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan; Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan; Nano Science and Technology Program, Taiwan International Graduate Program, Academia Sinica and National Taiwan University, Taiwan
| | - Ching-Chung Chang
- Doctoral Program in Tissue Engineering and Regenerative Medicine, National Chung-Hsing University, Taichung, Taiwan
| | | | - Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Hsien-Ming Lee
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan.
| | - Bill Cheng
- Graduate Institute of Biomedical Engineering, National Chung-Hsing University, Taichung, Taiwan.
| |
Collapse
|
7
|
Nabar SJ, Shet T, Malpani B, Hassan P, Basu S. Technetium-99m-labeled stealth liposomes: A new Strategy to Identify metastasis in tumour model. Appl Radiat Isot 2025; 218:111709. [PMID: 39923339 DOI: 10.1016/j.apradiso.2025.111709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVE Stealth liposomes are useful carriers for delivering drugs to cancer sites. In this paper we describe the preparation and evaluation of 99mTc labelled stealth liposomes (PEG-Liposomes) as potential radiopharmaceuticals for SPECT imaging of cancer. This study is first to describe targeted localisation of radiolabeled stealth liposomes in tumourous cells, sparing normal cells of various organs in metastatic L1210 mouse tumour model in BDF1 mice. METHODS Glutathione encapsulated stealth liposomes were made using lipid film hydration technique followed by probe sonication. Liposomes were radiolabeled using a lipophilic technetium complex, 99mTc-d,l-HMPAO. Labelled liposomes were purified by gel chromatography over sephadex G25 column. The tumour model was developed in immuno-competent BDF1 mice (F1DBA/2/C57-BL6 crosses) by injecting L1210 cells intraperitoneally. Biodistribution studies of the 99mTc- stealth liposomes were done in both tumour induced and normal mice. Histopathologic studies were done by excising the organs and the radioactivity in various sections was detected by autoradiography. RESULTS The stealth liposomes could be synthesized as per a reported procedure and it showed similar retention factor in thin layer chromatography. Liposomes could be radiolabelled by using 99mTc-d,lHMPAO. Purification over sephadex G 25 column yielded radioachemical purity greater than 95%. Biodistribution studies and autoradiography studies showed significantly higher accumulation of 99mTc labelled stealth liposomes in liver, pancreas and ascitic fluid of the tumour induced mice as compared to normal mice. CONCLUSIONS 99mTc labelled stealth liposomes having radiochemical purity greater than 95% could be prepared which showed higher uptake in tumour.
Collapse
Affiliation(s)
- Swapna J Nabar
- Homi Bhabha National Institute and Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Campus, Annex Building, JerbaiWadia Road, Dadar East, Mumbai, 400012, India.
| | - Tanuja Shet
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
| | - Basant Malpani
- Homi Bhabha National Institute and Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Campus, Annex Building, JerbaiWadia Road, Dadar East, Mumbai, 400012, India
| | - Puthusserickal Hassan
- Homi Bhabha National Institute, Bhabha Atomic Research Centre, Trombay, Mumbai, 400012, India
| | - Sandip Basu
- Homi Bhabha National Institute and Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Campus, Annex Building, JerbaiWadia Road, Dadar East, Mumbai, 400012, India
| |
Collapse
|
8
|
Menachem R, Nudelman I, Vorontsova A, Livneh I, Sela M, Benguigui M, Manobla B, Shammai Y, Deo A, Buxbaum C, Bessler R, Raviv Z, Shklover J, Sznitman J, Ciechanover A, Schroeder A, Shaked Y. Bone Marrow-Targeted Liposomes Loaded with Bortezomib Overcome Multiple Myeloma Resistance. ACS NANO 2025; 19:11684-11701. [PMID: 40117329 PMCID: PMC11966756 DOI: 10.1021/acsnano.4c10597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025]
Abstract
Multiple myeloma (MM) poses a significant therapeutic challenge due to its persistent progression and low survival rate. Although the proteasome inhibitor bortezomib has revolutionized MM treatment, MM aggressiveness and drug resistance remain critical concerns. To tackle this problem, we developed AMD3100-targeted Bortezomib Liposomes (ATBL) designed for the targeted delivery of bortezomib to MM cells. Uptake of ATBL into MM cells was dependent on CXCR4 and was enhanced compared to nontargeted liposomes, both in vitro and in vivo. Treating MM-bearing mice with ATBL achieved superior therapeutic efficacy compared to treatment with free bortezomib or nontargeted bortezomib-loaded liposomes. Notably, the therapeutic activity of ATBL was limited in mice inoculated with CXCR4-knockdown MM cells, highlighting CXCR4 as a potential biomarker for ATBL response. Importantly, ATBL was effective against an aggressive and bortezomib-resistant MM clone both in vitro and in vivo. Toxicity and biodistribution profiles demonstrated the safety and bone marrow-targeting ability of ATBL. Collectively, this study highlights ATBL as a promising next-generation proteasome inhibitor-based therapy that incorporates bone marrow-targeting ability and sensitizing elements to overcome drug resistance in MM.
Collapse
Affiliation(s)
- Rotem Menachem
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Igor Nudelman
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Avital Vorontsova
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Ido Livneh
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Mor Sela
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Madeleine Benguigui
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Bar Manobla
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Yael Shammai
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Abhilash Deo
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Chen Buxbaum
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Ron Bessler
- Faculty
of Biomedical Engineering, Technion−Israel
Institute of Technology, Haifa 3200001, Israel
| | - Ziv Raviv
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Jeny Shklover
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Josué Sznitman
- Faculty
of Biomedical Engineering, Technion−Israel
Institute of Technology, Haifa 3200001, Israel
| | - Aaron Ciechanover
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| | - Avi Schroeder
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
- Faculty
of Chemical Engineering, Technion −
Israel Institute of Technology, Haifa 3200003, Israel
| | - Yuval Shaked
- Department
of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion−Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport
Technion Integrated Cancer Center, Technion
− Israel Institute of Technology, Haifa 3525422, Israel
| |
Collapse
|
9
|
Tang J, Karbhari N, Campian JL. Therapeutic Targets in Glioblastoma: Molecular Pathways, Emerging Strategies, and Future Directions. Cells 2025; 14:494. [PMID: 40214448 PMCID: PMC11988183 DOI: 10.3390/cells14070494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, characterized by rapid growth, invasive infiltration into surrounding brain tissue, and resistance to conventional therapies. Despite advancements in surgery, radiotherapy, and chemotherapy, median survival remains approximately 15 months, underscoring the urgent need for innovative treatments. Key considerations informing treatment development include oncogenic genetic and epigenetic alterations that may dually serve as therapeutic targets and facilitate treatment resistance. Various immunotherapeutic strategies have been explored and continue to be refined for their anti-tumor potential. Technical aspects of drug delivery and blood-brain barrier (BBB) penetration have been addressed through novel vehicles and techniques including the incorporation of nanotechnology. Molecular profiling has emerged as an important tool to individualize treatment where applicable, and to identify patient populations with the most drug sensitivity. The goal of this review is to describe the spectrum of potential GBM therapeutic targets, and to provide an overview of key trial outcomes. Altogether, the progress of clinical and preclinical work must be critically evaluated in order to develop therapies for GBM with the strongest therapeutic efficacy.
Collapse
Affiliation(s)
- Justin Tang
- Department of Biomedical Science, University of Guelph, Guelph, ON N1G 2W1, Canada
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| | - Nishika Karbhari
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| | - Jian L. Campian
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| |
Collapse
|
10
|
Ediriweera GR, Li M, Fletcher NL, Houston ZH, Ahamed M, Blakey I, Thurecht KJ. Harnessing nanoparticles and bioorthogonal chemistries for improving precision of nuclear medicine. Biomater Sci 2025. [PMID: 40135276 DOI: 10.1039/d4bm01387e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
The convergence of nanotechnology, radiopharmaceutical development and molecular imaging has unveiled exciting opportunities for the progress of innovative diagnostic and therapeutic strategies, paving the way for significant advancements in biomedical research, especially in relation to cancer. For example, the use of highly sensitive and quantitative nuclear imaging techniques including PET and SPECT, together with nanoparticles for tumour imaging and therapy has recently expanded rapidly. While the long circulating properties of many nanomaterials are beneficial for prodrug chemotherapy formulations, due to the constant decay processes involved in nuclear medicines, directly labelled materials result in prolonged systemic radiation exposure and reduced therapeutic indices due to the unfavourable target-to-background ratios. This is due to the tendency for long circulating nanomaterials to distribute within the blood to other organs, such as the liver and spleen. The recent integration of bioorthogonal chemistry with nanotechnology and molecular imaging/radiotherapy has revolutionized the field by allowing the decoupling of the targeting molecule (i.e. nanomaterial with a bioorthogonal tag) and the imaging/therapeutic radioisotope. In this way, the detection/therapeutic element can be administered as a secondary "chase" molecule that contains the bioorthogonal partner, thereby creating an avenue to improve therapeutic index and provide imaging and treatments with reduced risk. This review will provide an overview of the progress made thus far in the field of nuclear imaging and radiotherapy for cancer using the combination of nanomaterials and bioorthogonal chemistry. We also provide a critical evaluation of the challenges and opportunities for using these approaches to better understand disease and treatment mechanisms, with the potential for downstream clinical translation.
Collapse
Affiliation(s)
- Gayathri R Ediriweera
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
- ARC Research Hub for Advanced Manufacture of Targeted Radiopharmaceuticals, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Mengdie Li
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Nicholas L Fletcher
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
- ARC Research Hub for Advanced Manufacture of Targeted Radiopharmaceuticals, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zachary H Houston
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Muneer Ahamed
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Idriss Blakey
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
- ARC Research Hub for Advanced Manufacture of Targeted Radiopharmaceuticals, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
- ARC Research Hub for Advanced Manufacture of Targeted Radiopharmaceuticals, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
11
|
Sun G, Wu Y, Li J, Yang M, Xu H, Li Y, Tong P, Shao R, Liu Y, Kong X. Quercetin liposomes conjugated with hyaluronidase: An efficient drug delivery system to block pancreatic cancer. J Control Release 2025; 382:113642. [PMID: 40127723 DOI: 10.1016/j.jconrel.2025.113642] [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: 11/26/2024] [Revised: 02/03/2025] [Accepted: 03/15/2025] [Indexed: 03/26/2025]
Abstract
Pancreatic cancer characterized with intense hydraulic tissue in tumor extracellular matrix (ECM) resists most of chemotherapeutic drugs. Increased levels of hyaluronic acid (HA) represent the primary component of the hydraulic tissue, rendering tumors protective from drug targeting. Quercetin (Que), a natural flavonoid, has the ability to inhibit tumor cell growth in a number of cancers; however, its poor water solubility and low bioavailability largely limit its application in cancer therapy. Hence, we developed an efficient drug delivery system by encapsulation of Que. into liposomes and conjugation with hyaluronidase (HAase) at liposome surface, termed as HQL. In the presence of HAase, HQL were predominantly accumulated at tumor with enhanced permeability and retention effect. Treatment of xenografted tumor mice with HQL gave rise to suppressed tumor growth, while no toxic effects were observed in mice. HQL demonstrated the strong ability to inhibit cell proliferation, promote cell apoptosis, and induce arrest at G2/M cell cycle in pancreatic cancer lines, three-dimensional cultured cell spheroids and pancreatic ductal adenocarcinoma (PDAC)-derived organoids. Mechanistically, HQL downregulated expression of cell cycle-associated protein (CCNB1, CDK1 and PLK1) and cell apoptosis-associated factors PI3K/AKT and Bcl-2. In summary, HQL degraded HA in the tumor microenvironment to enhance nano-particle penetration and inhibited tumor cell growth, eliciting efficacy of anti-tumor therapy. Thereof, HQL may provide a novel efficient drug delivery approach for the adjuvant treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ge Sun
- Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Ying Wu
- Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Jiekai Li
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Mingjie Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hang Xu
- Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Yiping Li
- Department of Oncology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430065, China
| | - Peilin Tong
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rong Shao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China; Shanghai Key Laboratory of Biliary Tract Diseases, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Department of General Surgery, Jiading Branch, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China; Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China.
| | - Xianming Kong
- Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Shanghai University of Medicine & Health Sciences, Shanghai 201318, China.
| |
Collapse
|
12
|
Karimi M, Aslanabadi A, Atkinson B, Hojabri M, Munawwar A, Zareidoodeji R, Ray K, Habibzadeh P, Parlayan HNK, DeVico A, Heredia A, Abbasi A, Sajadi MM. Subcutaneous liposomal delivery improves monoclonal antibody pharmacokinetics in vivo. Acta Biomater 2025; 195:522-535. [PMID: 39965705 DOI: 10.1016/j.actbio.2025.02.035] [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: 07/31/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
Background Monoclonal antibodies (mAbs) effectively treat and prevent various diseases, but their clinical application is hindered by issues related to the route of administration and pharmacokinetics (PK). Intravenous (IV) administration is cumbersome, while subcutaneous (SC) administration is hampered by lower bioavailability and potential for immunogenicity. This study evaluated the efficacy of liposomal formulations in enhancing the subcutaneous (SC) delivery and PK of broadly neutralizing antibodies (bNAbs) directed against HIV. METHODS mAbs were encapsulated in liposomes with and without PEGylation. The liposomes were characterized for particle size, polydispersity index, zeta potential, and release. Thereafter, mice were injected with free mAbs or liposome-encapsulated mAbs, and PK was evaluated. RESULTS Liposomes exhibited sizes of 85-92 nm with negative surface charges. Encapsulation efficiencies were 61 % for PEGylated and 58 % for non-PEGylated liposomes. Stability testing over 16 weeks revealed that formulations remained stable at 4 °C but showed leakage at 37 C. Cytotoxicity assays confirmed that the liposomal formulations did not affect cell viability or induce apoptosis in HMEC-1 cells. In vivo, PK studies in humanized FcRn mice indicated that the PEGylated formulations generally had higher half-life, Cmax, AUC, and MRT, and lower CL values compared to their non-PEGylated formulations of the same injection type. Both liposomal formulations showed improvements in bioavailability and extended half-life compared to free mAbs administered via SC and IV routes. Compared to the gold standard of IV free mAb injection, SC injection of antibodies encapsulated in PEGylated liposome had up to 80 % higher bioavailability and 45 % extension of half-life. Compared to the SC free mAb injection, the differences were even more pronounced, with liposomal SC injection having up to 113 % higher bioavailability and 81 % extension of half-life. CONCLUSION Overall, liposomal encapsulation effectively protected SC injected mAbs from degradation, facilitated sustained release, and improved PK profiles, suggesting a promising strategy for enhancing the therapeutic potential of mAbs in conditions that need repeated injections. Future work should further optimize liposomal formulations to increase loading capacity, stability, and release kinetics. STATEMENT OF SIGNIFICANCE This study addresses a challenge in the administration of monoclonal antibodies (mAbs). Intravenous administration requires additional resources, including nursing staff, making it time-consuming and costly. Although subcutaneous (SC) administration offers a less expensive and more patient-friendly option, it suffers from lower bioavailability and potentially shorter half-life. In this study, we encapsulated mAbs in liposomal formulations specifically designed to enhance their pharmacokinetics by promoting efficient lymphatic transport. Compared with both SC and even IV administration of free antibodies, liposomal formulations of mAbs remarkably improve bioavailability and extend the half-life. This innovative approach combines the comfort of SC administration with enhanced pharmacokinetics, addressing the limitations of current SC delivery methods. Liposomal formulations have the ability to greatly improve SC mAb administration by reducing the amount of antibody needed to be administered, reducing the frequency of injections, and potentially protecting against immunogenicity.
Collapse
Affiliation(s)
- Maryam Karimi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Arash Aslanabadi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ben Atkinson
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mahsa Hojabri
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Arshi Munawwar
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Roza Zareidoodeji
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Krishanu Ray
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Parham Habibzadeh
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hanife Nur Karakoc Parlayan
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Infectious Diseases, Karadeniz Technical University School of Medicine, Trabzon, Turkey
| | - Anthony DeVico
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alonso Heredia
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Abdolrahim Abbasi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Mohammad M Sajadi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
13
|
Zhang W, Pei B, Zhou Y, Li H, Ma W, Zhou B, Zhou C, Jiang H, Ji X. Emerging Targets, Novel Directions, and Innovative Approaches in Thrombosis Therapy. Aging Dis 2025:AD.2024.1688. [PMID: 40153578 DOI: 10.14336/ad.2024.1688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/11/2025] [Indexed: 03/30/2025] Open
Abstract
In clinical practice, antiplatelet, anticoagulant and fibrinolytic drugs are the mainstay of thrombosis treatment, but their potential bleeding side effects limit their widespread use. Therefore, modifying these existing drugs or developing new therapies that mitigate bleeding risks while maintaining their efficacy and utilization is necessary. Since the critical role of platelets in thrombosis is closely related to their cell surface receptors, intracellular signaling pathways and metabolism, current research focuses on these three major classes of platelet targets to develop new antithrombotic drugs. The coagulation cascade has always been the main target of anticoagulant drugs, but since the role of molecules of the contact system is more critical in thrombosis than in hemostasis, molecules targeting the contact system, such as FXIa and FXIIa, have become the main direction of anticoagulant drug research at present. Moreover, since the inflammatory response has been found to be significantly associated with thrombosis in recent years, the development of drugs that target inflammatory pathways, such as inflammasome, has also become a hot topic. This article provides a detailed description of these targets or drug formulations that are currently being investigated, including their mode of action and antithrombotic efficiency, and also points out their existing shortcomings. Moreover, antithrombotic nanomedicines can achieve precise release of drugs, which can greatly improve the thrombolytic efficiency and reduce side effects. In conclusion, this review focuses on summarizing the current new targets and new methods of antithrombotic drug research, hoping to provide a little reference for future related research.
Collapse
Affiliation(s)
- Weiyue Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Baoqing Pei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Hui Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Xunming Ji
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, 100069, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| |
Collapse
|
14
|
Fobian SF, Amin M, Sacchetti A, Oei AL, Ten Hagen TLM. Investigating the delivery of PD-L1-targeted immunoliposomes in a dynamic cervical cancer-on-a-chip model. J Control Release 2025; 379:236-250. [PMID: 39798703 DOI: 10.1016/j.jconrel.2025.01.014] [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: 06/30/2024] [Revised: 11/14/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
The recent approval of pembrolizumab in recurrent or metastatic cervical cancer warrants further investigations into the usefulness of immunotherapies for more durable and less radical interventions. In this study, the targeting potential of anti-PD-L1-functionalized immunoliposomes was tested in a 3D in vitro cervical cancer-on-a-chip model. Immunolipsomes were synthesized and decorated externally with monovalent anti-PD-L1 Fab' fragments of commercially available atezolizumab. Cervical cancer cell lines with varying levels of PD-L1 expression were cultured as spheroids embedded in a collagen I matrix, and treated under flow of culture media. Flow cytometry and live-cell confocal imaging were used to measure the interactions and uptake of untargeted liposomes and immunoliposomes in this panel of cell lines. The immunoliposomes retained specific functionality regardless of protein corona formation in high serum environments. As such, spheroids expressing high levels of PD-L1 preferentially internalized immunoliposomes in a 3D environment with extracellular matrix present, while low PD-L1-expressing cell lines showed no preference for either formulation. Importantly, treatments performed in monolayer cultures (on plastic) showed no differences between immuno- and untargeted liposome uptake, including the way in which the endocytosed liposomes are trafficked subcellularly. This study demonstrates the importance of both active and passive accumulation strategies to achieve nanoparticle targeting. Immunoliposomes remain a promising platform for the development of targeted nanotherapies against cervical cancers. However, initial functional tests did not translate directly to biological performance and this should be kept in mind for future formulations. Furthermore, the in vitro model developed appeared useful for visualizing liposome uptake in a 3D, live tissue environment and represents a cost-effective and reproducible model for future studies.
Collapse
Affiliation(s)
- Seth-Frerich Fobian
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands; Department of Radiation Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Cancer Center Amsterdam (CCA), Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Mohamadreza Amin
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Andrea Sacchetti
- Department of Pathology, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Arlene L Oei
- Department of Radiation Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Cancer Center Amsterdam (CCA), Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands.
| |
Collapse
|
15
|
Hetta HF, Melhem T, Aljohani HM, Salama A, Ahmed R, Elfadil H, Alanazi FE, Ramadan YN, Battah B, Rottura M, Donadu MG. Beyond Conventional Antifungals: Combating Resistance Through Novel Therapeutic Pathways. Pharmaceuticals (Basel) 2025; 18:364. [PMID: 40143141 PMCID: PMC11944814 DOI: 10.3390/ph18030364] [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: 01/29/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
The rising burden of fungal infections presents a significant challenge to global healthcare, particularly with increasing antifungal resistance limiting treatment efficacy. Early detection and timely intervention remain critical, yet fungal pathogens employ diverse mechanisms to evade host immunity and develop resistance, undermining existing therapeutic options. Limited antifungal options and rising resistance necessitate novel treatment strategies. This review provides a comprehensive overview of conventional antifungal agents, their mechanisms of action, and emerging resistance pathways. Furthermore, it highlights recently approved and investigational antifungal compounds while evaluating innovative approaches such as nanotechnology, drug repurposing, and immunotherapy. Addressing antifungal resistance requires a multifaceted strategy that integrates novel therapeutics, enhanced diagnostic tools, and future research efforts to develop sustainable and effective treatment solutions.
Collapse
Affiliation(s)
- Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Tameem Melhem
- Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague, Czech Republic;
| | - Hashim M. Aljohani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Madina 41477, Saudi Arabia;
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Ayman Salama
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Rehab Ahmed
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Hassabelrasoul Elfadil
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Fawaz E. Alanazi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Yasmin N. Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt;
| | - Basem Battah
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Antioch Syrian Private University, Maaret Saidnaya 22734, Syria;
| | - Michelangelo Rottura
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Matthew Gavino Donadu
- Hospital Pharmacy, Giovanni Paolo II Hospital, ASL Gallura, 07026 Olbia, Italy
- Department of Medicine, Surgery and Pharmacy, Scuola di Specializzazione in Farmacia Ospedaliera, University of Sassari, 07100 Sassari, Italy
| |
Collapse
|
16
|
Maksymova L, Pilger YA, Nuhn L, Van Ginderachter JA. Nanobodies targeting the tumor microenvironment and their formulation as nanomedicines. Mol Cancer 2025; 24:65. [PMID: 40033293 DOI: 10.1186/s12943-025-02270-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Among the emerging strategies for cancer theranostics, nanomedicines offer significant promise in advancing both patients' diagnosis and treatment. In combination with nanobodies, nanomedicines can potentially enhance the precision and efficiency of drug or imaging agent delivery, addressing key limitations of current approaches, such as off-target toxicities. The development of nanomedicines will be further accelerated by the creation of smart nanoparticles, and their integration with immunotherapy. Obviously, the success of nano-immunotherapy will depend on a comprehensive understanding of the tumor microenvironment, including the complex interplay of mechanisms that drive cancer-mediated immunosuppression and immune escape. Hence, effective therapeutic targeting of the tumor microenvironment requires modulation of immune cell function, overcoming resistance mechanisms associated with stromal components or the extracellular matrix, and/or direct elimination of cancer cells. Identifying key molecules involved in cancer progression and drug resistance is, therefore, essential for developing effective therapies and diagnostic tools that can predict patient responses to treatment and monitor therapeutic outcomes. Current nanomedicines are being designed with careful consideration of factors such as the choice of carrier (e.g., biocompatibility, controlled cargo release) and targeting moiety. The unique properties of nanobodies make them an effective engineering tool to target biological molecules with high affinity and specificity. In this review, we focus on the latest applications of nanobodies for targeting various components of the tumor microenvironment for diagnostic and therapeutic purposes. We also explore the main types of nanoparticles used as a carrier for cancer immunotherapies, as well as the strategies for formulating nanoparticle-nanobody conjugates. Finally, we highlight how nanobody-nanoparticle formulations can enhance current nanomedicines.
Collapse
Affiliation(s)
- Liudmyla Maksymova
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Pleinlaan 2, Brussels, B-1050, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Yannick A Pilger
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Faculty of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg, 97070, Germany
| | - Lutz Nuhn
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Faculty of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg, 97070, Germany.
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Pleinlaan 2, Brussels, B-1050, Belgium.
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.
| |
Collapse
|
17
|
Bonechi C, Tamasi G, Donati A, Bisozzi F, Baglioni M, Andreassi M, Ietta F, Leone G, Magnani A, Rossi C. Cationic liposomes as carriers of natural compounds from plant extract. Biophys Chem 2025; 318:107381. [PMID: 39733688 DOI: 10.1016/j.bpc.2024.107381] [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: 09/09/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/31/2024]
Abstract
Lipid-based nanocarriers provide versatile platforms for the encapsulation and delivery of many different bioactive compounds to improve the solubility, stability and therapeutic efficacy of bioactive phyto-compounds. In this study, liposomes were used to load leaf extract of Coffea Arabica, which is known to be rich beneficial substances such as alkaloids, flavonoids, etc. The aim of this work is to optimize the valorization of agricultural wastes containing natural antioxidants. The physico-chemical properties of the liposomes loaded with chlorogenic acid or Coffea arabica leaf extract were evaluated. The average size of empty and loaded liposomes was found to range of 120-150 nm, which is consistent with the fact that the addition of chlorogenic acid or Coffea arabica leaf extract can change the average size of the vesicles without affecting the physicochemical properties of the lipid bilayer, which remain stable systems. A structural and morphological characterization as well as an evaluation of biological properties such as viability in normal human dermal fibroblasts, is also been carried out. The cationic liposomes show a good average size and low polydispersity index values, indicating that the liposomes tend to be monodisperse and therefore stable. In particular, DOPC/DOTAP liposomes generally have better properties than DOPC/DDAB liposomes for use as encapsulation systems for natural plant extracts.
Collapse
Affiliation(s)
- Claudia Bonechi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy.
| | - Gabriella Tamasi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy
| | - Alessandro Donati
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy.
| | - Flavia Bisozzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy
| | - Michele Baglioni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy
| | - Marco Andreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy
| | - Francesca Ietta
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Gemma Leone
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121 Firenze, Italy
| | - Agnese Magnani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121 Firenze, Italy
| | - Claudio Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy
| |
Collapse
|
18
|
Oktay AN, Celebi N, Ilbasmis-Tamer S. Investigation of flurbiprofen pharmacokinetics in rats following dermal administration of optimized cyclodextrin-based nanogel. Eur J Pharm Sci 2025; 206:107021. [PMID: 39827972 DOI: 10.1016/j.ejps.2025.107021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/30/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE The main purpose of this study was to optimize a cyclodextrin-based nanogel of flurbiprofen (FP) for prolonged dermal administration and evaluate its stability, in vitro release, ex vivo skin permeation, and in vivo pharmacokinetic profile. METHODS The nanogels were prepared via emulsification/solvent evaporation process and optimized through design of experiments. Optimal formulation was characterized via particle size (PS), polydispersity index (PDI), zeta potential (ZP), differential scanning calorimetry (DSC) and X-ray powder diffraction (XRPD), solubility, stability, in vitro release/ex vivo permeation studies and mathematical modeling, and pharmacokinetic studies conducted in rats. Results were compared to HPMC-based gel that was not nano-sized (i.e.FP-HPMC gel). RESULTS The PS, PDI and ZP values of optimal FP-loaded nanogel were 295.5 nm, 0.361 and -31.9 mV, respectively and it was stable for 12 months. In in vitro release studies, the flux from the optimal FP-loaded nanogel (96.3 µg/hcm2) was three times slower (i.e.more controlled) than that of the FP-HPMC gel (287 µg/hcm2); the permeability coefficient of the nanogel (0.015 cm/h) was slightly less than that of FP-HPMC gel (0.046 cm/h). Rat skin studies showed FP-loaded nanogel provided higher drug retention in the skin, compared to FP-HPMC gel. Mathematical modeling from rat skin permeation showed the Hixson-Crowell model was the best fitting model for FP-loaded nanogel, suggesting surface area of the nanogel is changing during the release process. In rat pharmacokinetic studies, the FB-loaded nanogel exhibited prolonged and flatter plasma profile than the FP-HPMC gel, consistent with the higher drug retention in the skin. CONCLUSION The optimized nanogel provided prolonged drug permeation and more sustained pharmacokinetic performance compared to FP-HPMC gel.
Collapse
Affiliation(s)
- Ayse Nur Oktay
- Gazi University/Faculty of Pharmacy/Department of Pharmaceutical Technology, Ankara, Turkey; University of Health Sciences/Gulhane Faculty of Pharmacy/Department of Pharmaceutical Technology, Ankara, Turkey.
| | - Nevin Celebi
- Gazi University/Faculty of Pharmacy/Department of Pharmaceutical Technology, Ankara, Turkey; Başkent University/Faculty of Pharmacy/Department of Pharmaceutical Technology, Ankara, Turkey
| | - Sibel Ilbasmis-Tamer
- Gazi University/Faculty of Pharmacy/Department of Pharmaceutical Technology, Ankara, Turkey
| |
Collapse
|
19
|
Selim HMRM, Gomaa FAM, Alshahrani MY, Morgan RN, Aboshanab KM. Phage therapeutic delivery methods and clinical trials for combating clinically relevant pathogens. Ther Deliv 2025; 16:247-269. [PMID: 39545771 PMCID: PMC11875505 DOI: 10.1080/20415990.2024.2426824] [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: 07/15/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
The ongoing global health crisis caused by multidrug-resistant (MDR) bacteria necessitates quick interventions to introduce new management strategies for MDR-associated infections and antimicrobial agents' resistance. Phage therapy emerges as an antibiotic substitute for its high specificity, efficacy, and safety profiles in treating MDR-associated infections. Various in vitro and in vivo studies denoted their eminent bactericidal and anti-biofilm potential. This review addresses the latest developments in phage therapy regarding their attack strategies, formulations, and administration routes. It additionally discusses and elaborates on the status of phage therapy undergoing clinical trials, and the challenges encountered in their usage, and explores prospects in phage therapy research and application.
Collapse
Affiliation(s)
- Heba Mohammed Refat M. Selim
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University (Girls), Cairo, Egypt
| | - Fatma Alzahraa M. Gomaa
- Department of Pharmacognosy and Medicinal Herbs, Faculty of Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia
| | - Mohammad Y. Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Radwa N. Morgan
- National Centre for Radiation Research and Technology (NCRRT), Drug Radiation Research Department, Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Khaled M. Aboshanab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University Technology MARA (UiTM), Bandar Puncak Alam, Malaysia
| |
Collapse
|
20
|
Abavisani M, Khoshrou A, Eshaghian S, Karav S, Sahebkar A. Overcoming antibiotic resistance: the potential and pitfalls of drug repurposing. J Drug Target 2025; 33:341-367. [PMID: 39485073 DOI: 10.1080/1061186x.2024.2424895] [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: 06/09/2024] [Revised: 10/18/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Since its emergence shortly after the discovery of penicillin, antibiotic resistance has escalated dramatically, posing a significant health threat and economic burden. Drug repositioning, or drug repurposing, involves identifying new therapeutic applications for existing drugs, utilising their established safety profiles and pharmacological data to swiftly provide effective treatments against resistant pathogens. Several drugs, including otilonium bromide, penfluridol, eltrombopag, ibuprofen, and ceritinib, have demonstrated potent antibacterial activity against multidrug-resistant (MDR) bacteria. These drugs can disrupt biofilms, damage bacterial membranes, and inhibit bacterial growth. The combination of repurposed drugs with conventional antibiotics can reduce the required dosage of individual drugs, mitigate side effects, and delay the development of resistance, making it a promising strategy against MDR bacteria such as Staphylococcus aureus, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Escherichia coli. Despite its promise, drug repurposing faces challenges such as potential off-target effects, toxicity, and regulatory and intellectual property issues, necessitating rigorous evaluations and strategic solutions. This article aims to explore the potential of drug repurposing as a strategy to combat antibiotic resistance, examining its benefits, challenges, and future prospects. We address the legal, economic, and practical challenges associated with repurposing existing drugs, highlight successful examples, and propose solutions to enhance the efficacy and viability of this approach in combating MDR bacterial infections.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Souzan Eshaghian
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Kozak A, Lavrih E, Mikhaylov G, Turk B, Vasiljeva O. Navigating the Clinical Landscape of Liposomal Therapeutics in Cancer Treatment. Pharmaceutics 2025; 17:276. [PMID: 40006643 PMCID: PMC11859495 DOI: 10.3390/pharmaceutics17020276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Liposome-based targeted drug delivery systems represent a significant advancement in pharmaceutical science, offering distinct advantages that enhance the efficacy and safety of various therapies. These versatile carriers can encapsulate both hydrophilic and hydrophobic drugs, making them particularly valuable in clinical settings. This review explores the critical role of liposomal formulations in improving drug pharmacokinetics and minimizing side effects, especially in oncology, where targeted delivery to tumor cells is essential. Outlining the properties of different types of liposomes, we focus on the effects of these properties on the liposomes' targeting and drug release capabilities through innovative surface modifications and describe the most common methods of liposome preparation and characterization. Furthermore, this review provides an in-depth analysis of the properties and composition of liposomal-based nanocarriers, with a unique focus on ongoing clinical trials and recently approved therapies. It offers a comprehensive overview of the latest advancements in pre-clinical research and highlights the critical progress in clinical development, offering insights into the clinical impact and regulatory approvals. Ultimately, this review underscores the transformative potential of liposomal nanocarriers in modern therapeutics, suggesting avenues for future innovations and clinical breakthroughs.
Collapse
Affiliation(s)
- Andreja Kozak
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
| | - Ernestina Lavrih
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
- Jožef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Georgy Mikhaylov
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Olga Vasiljeva
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; (A.K.); (E.L.); (G.M.); (B.T.)
| |
Collapse
|
22
|
Rahma MN, Suhandi C, Mohammed AFA, El-Rayyes A, Elamin KM, Sulastri E, Wathoni N. The Role and Advancement of Liposomes for Oral Diseases Therapy. Int J Nanomedicine 2025; 20:1865-1880. [PMID: 39975418 PMCID: PMC11837752 DOI: 10.2147/ijn.s492353] [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: 08/20/2024] [Accepted: 11/25/2024] [Indexed: 02/21/2025] Open
Abstract
As many as 48.0% of the global population suffers from disabilities caused by oral conditions. These conditions encompass dental caries, periodontal diseases, oral cancers, and other pathologies affecting the hard and soft tissues of the oral and maxillofacial regions. Topical drug treatments in the oral cavity are often ineffective due to the short contact time, which prevents the drug from reaching optimal concentrations necessary for therapeutic effect. Conventional liposomes have several limitations, including low stability, challenges in long-term storage, and rapid clearance by the reticuloendothelial system (RES). These factors significantly reduce their effectiveness in maintaining sustained drug delivery and achieving desired therapeutic outcomes. To overcome these challenges, advanced drug delivery systems have been developed. Among these systems, liposomes have been extensively explored as nanocarriers in targeted drug delivery systems, particularly in mucosal drug delivery, due to their biocompatibility and degradability, making them promising agents for the treatment of oral diseases. To address these issues, extensive research has been conducted to modify the surface of liposomes, optimizing their efficacy, and understanding their mechanisms of action. This review article discusses the role and recent advancements of liposomes in the treatment of oral diseases, highlighting their potential to revolutionize oral health care through improved drug delivery and therapeutic outcomes.
Collapse
Affiliation(s)
- Maya Nurul Rahma
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| | - Ahmed F A Mohammed
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Ali El-Rayyes
- Center for Scientific Research and Entrepreneurship, Northern Border University, Arar, 73213, Saudi Arabia
| | - Khaled M Elamin
- Graduated School of Pharmaceutical Science, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Evi Sulastri
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Tadulako University, Palu, Central Sulawesi, 94118, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang, West Java, 45363, Indonesia
| |
Collapse
|
23
|
Arandhara A, Bhuyan P, Das BK. Exploring lung cancer microenvironment: pathways and nanoparticle-based therapies. Discov Oncol 2025; 16:159. [PMID: 39934547 DOI: 10.1007/s12672-025-01902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Lung cancer stands out as a significant global health burden, with staggering incidence and mortality rates primarily linked to smoking and environmental carcinogens. The tumor microenvironment (TME) emerges as a critical determinant of cancer progression and treatment outcomes, comprising a complex interplay of cells, signaling molecules, and extracellular matrix. Through a comprehensive literature review, we elucidate current research trends and therapeutic prospects, aiming to advance our understanding of TME modulation strategies and their clinical implications for lung cancer treatment. Dysregulated immune responses within the TME can facilitate tumor evasion, limiting the efficacy of immune checkpoint inhibitors (ICI). Consequently, TME modulation strategies have become potential avenues to enhance therapeutic responses. However, conventional TME-targeted therapies often face challenges. In contrast, nanoparticle (NP)-based therapies offer promising prospects for improved drug delivery and reduced toxicity, leveraging the enhanced permeability and retention (EPR) effect. Despite NP design and delivery advancements, obstacles like poor tumor cell uptake and off-target effects persist, necessitating further optimization. This review underscores the pivotal role of TME in lung cancer management, emphasizing the synergistic potential of immunotherapy and nano-therapy.
Collapse
Affiliation(s)
- Arunabh Arandhara
- Assam Pharmacy Institute, Titabar, Amgurikhat, Jorhat, Assam, 785632, India
| | - Pallabi Bhuyan
- School of Pharmacy, The Assam Kaziranga University, Koraikhowa, Jorhat, Assam, 785006, India
| | - Bhrigu Kumar Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam, 781017, India.
| |
Collapse
|
24
|
Ha H, Choi Y, Kim NH, Kim J, Jang J, Niepa THR, Tanaka M, Lee HY, Choi J. Lipid Nanoparticle Delivery System for Normalization of Tumor Microenvironment and Tumor Vascular Structure. Biomater Res 2025; 29:0144. [PMID: 39935791 PMCID: PMC11811622 DOI: 10.34133/bmr.0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/07/2025] [Accepted: 01/18/2025] [Indexed: 02/13/2025] Open
Abstract
Tumors grow by receiving oxygen and nutrients from the surrounding blood vessels, leading to rapid angiogenesis. This results in functionally and structurally abnormal vasculature characterized by high permeability and irregular blood flow, causing hypoxia within the tumor microenvironment (TME). Hypoxia exacerbates the secretion of pro-angiogenic factors such as vascular endothelial growth factor (VEGF), further perpetuating abnormal vessel formation. This environment compromises the efficacy of radiotherapy, immunotherapy, and chemotherapy. In this study, we developed a pH-sensitive liposome (PSL) system, termed OD_PSL@AKB, to co-deliver oxygen (OD) and razuprotafib (AKB-9778) to tumors. This system rapidly responds to the acidic TME to alleviate hypoxia and inhibit VEGF secretion, restoring VE-cadherin expression in hypoxic endothelial cell/cancer cell cocultures. Our findings highlight the potential of OD_PSL@AKB in normalizing tumor vasculature and improving therapeutic efficacy.
Collapse
Affiliation(s)
- Heejin Ha
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Department of Chemical Science and Engineering,
Institute of Science Tokyo, Kanagawa 226-8503, Japan
| | - Na-Hyeon Kim
- Department of Chemical Engineering,
Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Jiwon Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jaehee Jang
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Tagbo H. R. Niepa
- Department of Chemical Engineering,
Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Biomedical Engineering,
Carnegie Mellon University, Pittsburgh, PA, USA
| | - Masayoshi Tanaka
- Department of Chemical Science and Engineering,
Institute of Science Tokyo, Kanagawa 226-8503, Japan
| | - Hee-Young Lee
- Department of Chemical Engineering,
Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| |
Collapse
|
25
|
Song Y, Li N, Luo Q, Liu D, Wang Z. Intranasal Administrations of AP39-Loaded Liposomes Selectively Deliver H2S to Neuronal Mitochondria to Protect Neonatal Hypoxia-Ischemia by Targeting ERK1/2 and Caspase-1. ACS Biomater Sci Eng 2025; 11:1184-1197. [PMID: 39841892 DOI: 10.1021/acsbiomaterials.4c02282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Mitochondrial dysfunction contributes to the pathology of hypoxia-ischemia (HI) brain damage by aberrant production of ROS. Hydrogen sulfide (H2S) has been demonstrated to exert neuroprotective effects through antioxidant mechanisms. However, the diffusion of H2S in vivo is not specifically targeted and may even be systemically toxic. In this study, based on mitochondria-targeted H2S donor AP39, we fabricated liposomes encapsulating AP39 (AP39@Lip) via intranasal delivery to improve functional recovery after HI brain injury. This study presents that intranasal administration of AP39@Lip was capable of attenuating acute brain injury by inhibiting mitochondrial dysfunction, apoptosis, neuroinflammation, and ROS production in the lesional cortex 3 days after HI brain injury. Similarly, AP39@Lip was observed to restore both short- and long-term function following HI injury without obvious toxicity. Mechanistically, the therapeutic effects of AP39@Lip mainly relied on its colocalization with neuronal mitochondria 24 h after administration and reversed H2S levels in the lesional cortex. Moreover, molecular docking and cellular thermal shift assay suggest that AP39 inhibited the activation of ERK1/2 and caspase-1 by directly binding to ERK1/2 or caspase-1. These results indicate that intranasal administration of AP39@Lip selectively delivered H2S to neuronal mitochondria and mitigated mitochondrial damage following HI insult by targeting ERK1/2 and caspase-1.
Collapse
Affiliation(s)
- Yan Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Nianlu Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, P.R. China
| | - Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
26
|
Alfei S, Zuccari G. Last Fifteen Years of Nanotechnology Application with Our Contribute. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:265. [PMID: 39997828 PMCID: PMC11858446 DOI: 10.3390/nano15040265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025]
Abstract
Currently, nanotechnology is the most promising science, engineering, and technology conducted at the nanoscale (nm), which is used in several sectors. Collectively, nanotechnology is causing a new industrial revolution, and nano-based products are becoming increasingly important for the global market and economy. The interest in nanomaterials has been strongly augmented during the last two decades, and this fact can be easily evaluated by considering the number of studies present in the literature. In November 2024, they accounted for 764,279 experimental studies developed in the years 2009-2024. During such a period, our group contributed to the field of applicative nanotechnology with several experimental and review articles, which we hope could have relevantly enhanced the knowledge of the scientific community. In this new publication, an exhaustive overview regarding the main types of developed nanomaterials, the characterization techniques, and their applications has been discussed. Particular attention has been paid to nanomaterials employed for the enhancement of bioavailability and delivery of bioactive molecules and to those used for ameliorating traditional food packaging. Then, we briefly reviewed our experimental studies on the development of nanoparticles (NPs), dendrimers, micelles, and liposomes for biomedical applications by collecting inherent details in a reader-friendly table. A brief excursus about our reviews on the topic has also been provided, followed by the stinging question of nanotoxicology. Indeed, although the application of nanotechnology translates into a great improvement in the properties of non-nanosized pristine materials, there may still be a not totally predictable risk for humans, animals, and the environment associated with an extensive application of NPs. Nanotoxicology is a science in rapid expansion, but several sneaky risks are not yet fully disclosed. So, the final part of this study discusses the pending issue related to the possible toxic effects of NPs and their impact on customers' acceptance in a scenario of limited knowledge.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy (DIFAR), University of Genoa, Via Cembrano 4, 16148 Genoa, Italy;
| | - Guendalina Zuccari
- Department of Pharmacy (DIFAR), University of Genoa, Via Cembrano 4, 16148 Genoa, Italy;
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genoa, Italy
| |
Collapse
|
27
|
Liu L, He H, Du B, He Y. Nanoscale drug formulations for the treatment of Alzheimer's disease progression. RSC Adv 2025; 15:4031-4078. [PMID: 39926227 PMCID: PMC11803502 DOI: 10.1039/d4ra08128e] [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: 11/16/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder with no effective disease-modifying treatments. The blood-brain barrier hinders drug delivery to the brain, limiting therapeutic efficacy. Nanoparticle-based systems have emerged as promising tools to overcome these challenges. This review highlights recent advances in nanoparticle technologies for AD treatment, including liposomes, polymeric, inorganic, and biomimetic nanoparticles. These nanoparticles improve drug delivery across the blood-brain barrier, improve stability and bioavailability, and enable targeted delivery to affected brain regions. Functionalization strategies further enhance their therapeutic potential. Multifunctional nanoparticles combining therapeutic and diagnostic properties offer theranostic approaches. While progress has been made, challenges related to safety, targeting precision, and clinical translation remain. Future perspectives emphasize the need for collaborative efforts to optimize nanoparticle design, conduct rigorous studies, and accelerate the development of effective nanotherapeutics. With continued innovation, nanoparticle-based delivery systems hold great promise for revolutionizing AD treatment.
Collapse
Affiliation(s)
- Liqin Liu
- Department of Pediatrics of Neurology Nursing, West China School of Nursing, West China Second University Hospital, Sichuan University Chengdu 610000 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu 610000 China
| | - Haini He
- Department of Pediatrics of Neurology Nursing, West China School of Nursing, West China Second University Hospital, Sichuan University Chengdu 610000 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu 610000 China
| | - Bin Du
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610000 China
| | - Yang He
- Department of Pediatrics, West China Second University Hospital, Sichuan University Chengdu 610000 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu 610000 China
| |
Collapse
|
28
|
Li Z, Xi Z, Fan C, Xi X, Zhou Y, Zhao M, Xu L. Nanomaterials evoke pyroptosis boosting cancer immunotherapy. Acta Pharm Sin B 2025; 15:852-875. [PMID: 40177577 PMCID: PMC11959974 DOI: 10.1016/j.apsb.2024.11.011] [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: 07/10/2024] [Revised: 09/12/2024] [Accepted: 10/15/2024] [Indexed: 04/05/2025] Open
Abstract
Cancer immunotherapy is currently a very promising therapeutic strategy for treating tumors. However, its effectiveness is restricted by insufficient antigenicity and an immunosuppressive tumor microenvironment (ITME). Pyroptosis, a unique form of programmed cell death (PCD), causes cells to swell and rupture, releasing pro-inflammatory factors that can enhance immunogenicity and remodel the ITME. Nanomaterials, with their distinct advantages and different techniques, are increasingly popular, and nanomaterial-based delivery systems demonstrate significant potential to potentiate, enable, and augment pyroptosis. This review summarizes and discusses the emerging field of nanomaterials-induced pyroptosis, focusing on the mechanisms of nanomaterials-induced pyroptosis pathways and strategies to activate or enhance specific pyroptosis. Additionally, we provide perspectives on the development of this field, aiming to accelerate its further clinical transition.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ziyue Xi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chuanyong Fan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinran Xi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yao Zhou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ming Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
29
|
Prabhu C, Satyaprasad AU, Deekshit VK. Understanding Bacterial Resistance to Heavy Metals and Nanoparticles: Mechanisms, Implications, and Challenges. J Basic Microbiol 2025; 65:e2400596. [PMID: 39696916 DOI: 10.1002/jobm.202400596] [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: 09/10/2024] [Revised: 10/30/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
Antimicrobial resistance is a global health problem as it contributes to high mortality rates in several infectious diseases. To address this issue, engineered nanoparticles/nano-formulations of antibiotics have emerged as a promising strategy. Nanoparticles are typically defined as materials with dimensions up to 100 nm and are made of different materials such as inorganic particles, lipids, polymers, etc. They are widely dispersed in the environment through various consumer products, and their clinical applications are diverse, ranging from contrast agents in imaging to carriers for gene and drug delivery. Nanoparticles can also act as antimicrobial agents either on their own or in combination with traditional antibiotics to produce synergistic effects, earning them the label of "next-generation therapeutics." They have also shown great effectiveness against multidrug-resistant pathogens responsible for nosocomial infections. However, overexposure or prolonged exposure to sublethal doses of nanoparticles can promote the development of resistance in human pathogens. The resistance can arise from various factors such as genetic mutation, horizontal gene transfer, production of reactive oxygen species, changes in the outer membrane of bacteria, efflux-induced resistance, cross-resistance from intrinsic antibiotic resistance determinants, plasmid-mediated resistance, and many more. Continuous exposure to nanoparticles can also transform an antibiotic-susceptible bacterial pathogen into multidrug resistance. Considering all these, the current review focuses on the mode of action of different heavy metals and nanoparticles and possible mechanisms through which bacteria attain resistance towards these heavy metals and nanoparticles.
Collapse
Affiliation(s)
- Chaitra Prabhu
- Department of Infectious Diseases and Microbial Genomics, NITTE (Deemed to be University), NITTE University Centre for Science Education and Research, Paneer Campus, Deralakatte, Mangaluru, India
| | - Akshath Uchangi Satyaprasad
- Department of Bio and Nano Technology, NITTE (Deemed to be University), NITTE University Centre for Science Education and Research, Paneer Campus, Deralakatte, Mangaluru, India
| | - Vijaya Kumar Deekshit
- Department of Infectious Diseases and Microbial Genomics, NITTE (Deemed to be University), NITTE University Centre for Science Education and Research, Paneer Campus, Deralakatte, Mangaluru, India
| |
Collapse
|
30
|
Xiong T, Li D, Ren J, Chen C, Li S, Song Z, Xu N, Liu T, Liu S. Soluble microneedle acupuncture patches containing melittin liposomes for the percutaneous treatment of rheumatoid arthritis. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 64:102806. [PMID: 39855442 DOI: 10.1016/j.nano.2025.102806] [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: 03/22/2024] [Revised: 05/11/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025]
Abstract
Bee venom acupuncture (BVA) offers therapeutic potential for rheumatoid arthritis (RA) but faces challenges from pain and allergies linked to live bee stings. A key hurdle is melittin (Mel), bee venom's main anti-inflammatory component, which degrades rapidly when orally ingested, leading to decreased efficacy and increased toxicity. This study proposes a solution by encapsulating melittin in liposomes to enhance stability and lessen side effects, expanding its clinical applicability. Additionally, the advancement of microneedle technology, which bypasses gastrointestinal issues by targeting the stratum corneum, opens a novel pathway for RA treatment. Employing soluble microneedles loaded with melittin-encapsulated liposomes (Mel-Lip) enables effective transdermal delivery. Results from an adjuvant-induced RA animal model show that Mel-Lip microneedles improve foot health, repair cartilage, and lower inflammatory markers, highlighting microneedling with transdermal nanocarriers as a promising, patient-friendly approach for RA management.
Collapse
Affiliation(s)
- Tong Xiong
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Du Li
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Juanjuan Ren
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Chuncheng Chen
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Shijie Li
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Zhuoyue Song
- Bioengineering Laboratory, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510070, Guangdong, PR China
| | - Nenggui Xu
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Tao Liu
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Shihui Liu
- Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| |
Collapse
|
31
|
Simon L, Constanzo J, Terraza-Aguirre C, Ibn Elfekih Z, Berthelot J, Benkhaled BT, Haute T, Pednekar K, Clark K, Emerson SJ, Atis S, Benedetti C, Langlois S, Marquant A, Prakash J, Wang A, Devoisselle JM, Montier T, Djouad F, Pouget JP, Lapinte V, Morille M. Surface modification of extracellular vesicles with polyoxazolines to enhance their plasma stability and tumor accumulation. Biomaterials 2025; 313:122748. [PMID: 39180918 DOI: 10.1016/j.biomaterials.2024.122748] [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: 04/26/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Extracellular vesicles (EVs) are future promising therapeutics, but their instability in vivo after administration remains an important barrier to their further development. Many groups evaluated EV surface modification strategies to add a targeting group with the aim of controlling EV biodistribution. Conversely, fewer groups focused on their stabilization to obtain "stealth" allogenic EVs. Modulating their stabilization and biodistribution is an essential prerequisite for their development as nano-therapeutics. Here, we explored polyoxazolines with lipid anchors association to the EV membrane (POxylation as an alternative to PEGylation) to stabilize EVs in plasma and control their biodistribution, while preserving their native properties. We found that this modification maintained and seemed to potentiate the immunomodulatory properties of EVs derived from mesenchymal stem/stromal cells (MSC). Using a radiolabeling protocol to track EVs at a therapeutically relevant concentration in vivo, we demonstrated that POxylation is a promising option to stabilize EVs in plasma because it increased EV half-life by 6 fold at 6 h post-injection. Moreover, EV accumulation in tumors was higher after POxylation than after PEGylation.
Collapse
Affiliation(s)
- L Simon
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | | | - Z Ibn Elfekih
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Berthelot
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - B T Benkhaled
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Haute
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - K Pednekar
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - K Clark
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S J Emerson
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S Atis
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - C Benedetti
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - S Langlois
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - A Marquant
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Prakash
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - A Wang
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - J M Devoisselle
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de La Reproduction, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200, Brest, France
| | - F Djouad
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France
| | - J P Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - V Lapinte
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - Marie Morille
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France; Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
32
|
Emami J, Kazemi M, Mirian M. Synthesis and in vitro evaluation of self-assembling biocompatible heparin-based targeting polymeric micelles for delivery of doxorubicin to leukemic cells. Res Pharm Sci 2025; 20:142-164. [PMID: 40190826 PMCID: PMC11972022 DOI: 10.4103/rps.rps_197_24] [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: 10/21/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 04/09/2025] Open
Abstract
Background and purpose Biodegradable polymeric micelles have emerged as one of the most promising platforms for targeted drug delivery. In the present study, a polymeric micelle composed of folic acid (FA), heparin (HEP), dexamethasone (DEX), and (FA-PEG-HEP-CA-TOC) was developed for the delivery of doxorubicin (DOX) to leukemic cells. Experimental approach FA-HEP-DEX was synthesized and characterized by 1H-NMR. DOX-loaded micelles were prepared using a dialysis method. The impact of various processing variables, including polymer-to-drug ratio, dialysis temperature, and solvent type, on the physicochemical properties of the micelles were evaluated. In vitro, cellular uptake and cytotoxicity of the micelles in folate receptor-positive (K562) and negative (HepG2) cells were evaluated. Findings/Results The 1H-NMR results confirmed the successful synthesis of FA-HEP-DEX. DOX-loaded micelles exhibited an average particle size of 117 to 181 nm with a high drug entrapment efficiency (36% to 71%). DOX-loaded micelles also showed sustained drug-release behavior. DOX-loaded FA-HEP-DEX micelles exhibited higher cellular uptake and in vitro cytotoxicity than free DOX and DOX-loaded HEP-DEX micelles in K562 cells. Conclusions and implications DOX was well incorporated into the micelles with high entrapment efficiency due to high solubility of DOX in DEX as the hydrophobic component of the micelle structure. The higher cellular uptake and cell toxicity of targeted micelles correspond to the presence of FA on the micelle surface, which promotes cell internalization of the micelles viaspecific receptor-mediated endocytosis. Our results indicated the potential of DOX-loaded heparin-based micelles with desirable antitumor activity as a targeted drug delivery system in cancer therapy.
Collapse
Affiliation(s)
- Jaber Emami
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Moloud Kazemi
- Nanotechnology Research Center, Medical Basic Research Sciences Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
33
|
Chen J, Zhao Z, Alantary D, Huang J. Nanomedicine for pediatric healthcare: A review of the current state and future prospectives. Eur J Pharm Biopharm 2025; 207:114597. [PMID: 39647671 DOI: 10.1016/j.ejpb.2024.114597] [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: 07/07/2024] [Revised: 11/10/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
Nanomedicine has emerged as a valuable treatment and diagnosis option, due to its ability not only to address formulation challenges associated with new therapeutic moieties, but also to improve the existing drugs efficacy. Nanomedicine provides appealing advantages such as increased drug payload, enhanced stability, tailored drug release profile, improved bioavailability and targeted drug delivery, etc. Tremendous research and regulatory efforts have been made in the past decades to advance nanomedicine from the benchtop to clinic. Numerous nanotechnology-based formulation approaches have been seen succeeding in commercialization. Despite the progress in nanomedicine use in adults, the advancement in pediatric population has been much slower. Clearly the treatment of disease in children cannot be simplified by dose adjustment based on body weight or surface, due to the significant differences in physiology thus the drug absorption, distribution, metabolism, excretion and transport (ADMET), between children and adults. This inherent variable among others poses much more challenges when developing pediatric-specific nanomedicine or translating adult nanodrug to pediatric indication. This review therefore intends to highlight the physiological differences between children and adult, and the common pediatric diseases which are good candidates for nanomedicine. The formulation approaches utilized in the marketed nanomedicine with pediatric indications, including liposomes, nanocrystals, polymeric nanoparticles and lipid nanoemulsions are elaborated. Finally, the challenges and gaps in pediatric nanomedicine development and commercialization, and the future prospectives are discussed.
Collapse
Affiliation(s)
- Jiayi Chen
- Ascendia Pharmaceuticals, Inc., North Brunswick, NJ 08902, United States
| | - Zhifeng Zhao
- Ascendia Pharmaceuticals, Inc., North Brunswick, NJ 08902, United States
| | - Doaa Alantary
- Ascendia Pharmaceuticals, Inc., North Brunswick, NJ 08902, United States
| | - Jingjun Huang
- Ascendia Pharmaceuticals, Inc., North Brunswick, NJ 08902, United States.
| |
Collapse
|
34
|
Guedes M, Vieira de Castro J, Lima AC, M F Gonçalves V, Tiritan ME, L Reis R, Ferreira H, M Neves N. Fishroesomes show intrinsic anti-inflammatory bioactivity and ability as celecoxib carriers in vivo. Eur J Pharm Biopharm 2025; 207:114587. [PMID: 39645203 DOI: 10.1016/j.ejpb.2024.114587] [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: 04/24/2024] [Revised: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
According to the World Health Organization (WHO), chronic inflammatory-related diseases represent the greatest threat to human health. Indeed, failure in the resolution of inflammation leads to serious pathological conditions, such as cardiovascular diseases, arthritis, cancer, diabetes, autoimmune diseases, and neurodegenerative disorders that are often associated with extremely high human suffering and societal and economic burdens. Despite the number and efficacy of available therapeutic agents have been increased, the serious side effects associated with some of them often create a very high risk/benefit ratio for patients. Therefore, herein, a drug delivery system was engineered to overcome important drawbacks of conventional therapies and to have a synergistic action with the incorporated drug. Indeed, it will have an added beneficial role in controlling inflammation. For that, sardine (Sardina pilchardus) roe was used as the lipidic source to produce bioactive liposomes, namely fishroesomes. These spherical vesicles with ≈326 nm in size and a significant negative surface charge (≈-31 mV) were able to encapsulate and control the release of the anti-inflammatory drug celecoxib. Moreover, fishroesomes were cytocompatible for different cell types (chondrocytes and macrophages), at concentrations in which they present anti-inflammatory properties. Importantly, fishroesomes were more effective in reducing pro-inflammatory mediators than the free drug. We also demonstrated that a single intra-articular injection of the fishroesomes encapsulating or not celecoxib in an experimental rat model of inflammatory arthritis was safe and more effective in controlling the pain and reducing the synovial inflammation compared to the free drug. Notably, as the celecoxib concentration in the sardine roe-derived liposomes was less than half of the amount of free drug, this study demonstrates the value of fishroesomes in counteracting inflammation. Therefore, the developed formulations may be considered a promising therapeutic option for inflammatory conditions.
Collapse
Affiliation(s)
- Marta Guedes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Vieira de Castro
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Cláudia Lima
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Virgínia M F Gonçalves
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal
| | - Maria Elizabeth Tiritan
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal; Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal; Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia da Universidade do Porto, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Helena Ferreira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Nuno M Neves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Rua Ave 1, Edifício 1 (Sede), 4805-694 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
35
|
Izadiyan Z, Misran M, Kalantari K, Webster TJ, Kia P, Basrowi NA, Rasouli E, Shameli K. Advancements in Liposomal Nanomedicines: Innovative Formulations, Therapeutic Applications, and Future Directions in Precision Medicine. Int J Nanomedicine 2025; 20:1213-1262. [PMID: 39911259 PMCID: PMC11794392 DOI: 10.2147/ijn.s488961] [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: 07/27/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
Liposomal nanomedicines have emerged as a pivotal approach for the treatment of various diseases, notably cancer and infectious diseases. This manuscript provides an in-depth review of recent advancements in liposomal formulations, highlighting their composition, targeted delivery strategies, and mechanisms of action. We explore the evolution of liposomal products currently in clinical trials, emphasizing their potential in addressing diverse medical challenges. The integration of immunotherapeutic agents within liposomes marks a paradigm shift, enabling the design of 'immuno-modulatory hubs' capable of orchestrating precise immune responses while facilitating theranostic applications. The recent COVID-19 pandemic has accelerated research in liposomal-based vaccines and antiviral therapies, underscoring the need for improved delivery mechanisms to overcome challenges like rapid clearance and organ toxicity. Furthermore, we discuss the potential of "smart" liposomes, which can respond to specific disease microenvironments, enhancing treatment efficacy and precision. The integration of artificial intelligence and machine learning in optimizing liposomal designs promises to revolutionize personalized medicine, paving the way for innovative strategies in disease detection and therapeutic interventions. This comprehensive review underscores the significance of ongoing research in liposomal technologies, with implications for future clinical applications and enhanced patient outcomes.
Collapse
Affiliation(s)
- Zahra Izadiyan
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Misni Misran
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Katayoon Kalantari
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Thomas J Webster
- Biomedical Engineering, Hebei University of Technology, Tianjin, People’s Republic of China
- School of Engineering, Saveetha University, Chennai, India
| | - Pooneh Kia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | - Elisa Rasouli
- Department of Electrical and Electronics Engineering, Nanyang Technological University, Nanyang, Singapore
| | - Kamyar Shameli
- School of Medicine, Institute of Virology, Technical University of Munich, Munich, Germany
| |
Collapse
|
36
|
Azimizonuzi H, Ghayourvahdat A, Ahmed MH, Kareem RA, Zrzor AJ, Mansoor AS, Athab ZH, Kalavi S. A state-of-the-art review of the recent advances of theranostic liposome hybrid nanoparticles in cancer treatment and diagnosis. Cancer Cell Int 2025; 25:26. [PMID: 39871316 PMCID: PMC11773959 DOI: 10.1186/s12935-024-03610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/10/2024] [Indexed: 01/29/2025] Open
Abstract
Theranostics is a way of treating illness that blends medicine with testing. Specific characteristics should be present in the best theranostic agents for cancer: (1) the drugs should be safe and non-toxic; (2) they should be able to treat cancer selectively; and (3) they should be able to build up only in the cancerous tissue. Liposomes (LPs) are one of the most efficient drug delivery methods based on nanotechnology. Stealth LPs and commercial LPs have recently had an impact on cancer treatment. Using the valuable information from each imaging technique, along with the multimodality imaging functionality of liposomal therapeutic agents, makes them very appealing for personalized monitoring of how well therapeutic drugs are working against cancer in vivo and for predicting how well therapies will work. On the other hand, their use as nanoparticle delivery systems is currently in the research and development phase. Nanoscale delivery system innovation has made LP-nanoparticle hybrid structures very useful for combining therapeutic and imaging methods. LP-hybrid nanoparticles are better at killing cancer cells than their LP counterparts, making them excellent options for in vivo and in vitro drug delivery applications. Hybrid liposomes (HLs) could be used in the future as theranostic carriers to find and treat cancer targets. This would combine the best features of synthetic and biological drug delivery systems. Overarchingly, this article provided a comprehensive overview of the many LP types used in cancer detection, therapy, and theranostic analysis. An evaluation of the pros and cons of the many HLs types used in cancer detection and treatment has also been conducted. The study also included recent and significant research on HLs for cancer theranostic applications. We conclude by outlining the potential benefits and drawbacks of this theranostic approach to the concurrent detection and treatment of different malignancies, as well as its prospects.
Collapse
Affiliation(s)
- Hannaneh Azimizonuzi
- Inventor Member of International Federation of Inventors Associations, Geneva, Switzerland
| | - Arman Ghayourvahdat
- Inventor Member of International Federation of Inventors Associations, Geneva, Switzerland
| | | | | | - Athmar Jaber Zrzor
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Shaylan Kalavi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Islamic Azad University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Alberto KA, Hasna Begam MN, Xiong H, Shinoda W, Slesinger PA, Qin Z, Nielsen SO. Fully atomistic molecular dynamics modeling of photoswitchable azo-PC lipid bilayers: structure, mechanical properties, and drug permeation. NANOSCALE 2025; 17:2032-2042. [PMID: 39641529 DOI: 10.1039/d4nr02509a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Phospholipid based vesicles called liposomes are commonly used as packaging in advanced drug delivery applications. Stimuli-responsive liposomes have been designed to release their contents under certain conditions, for example through heating or illumination. However, in the case of photosensitive liposomes based on azo-PC, namely phosphatidylcholine lipids with azobenzene incorporated into one of the two lipid tails, the release mechanism is not known. Here we show, using fully-atomistic molecular dynamics simulations of pure azo-PC bilayers, that drug permeation through the bilayer is driven by a light-induced gel-to-liquid lipid phase transition that softens the membrane bending rigidity by an order of magnitude, increases the area per lipid, and decreases the membrane thickness. Furthermore, using phenol as a model drug, we quantified its translocation free energy and its ability to cross the bilayer as a result of a chemical potential gradient induced through a double-bilayer simulation unit cell. The molecular level structural and dynamic information obtained in this study should be of help in designing new azo-PC based liposomes.
Collapse
Affiliation(s)
- Kevin A Alberto
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA.
| | - M N Hasna Begam
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Hejian Xiong
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wataru Shinoda
- Research Institute for Interdisciplinary Science, Okayama University, Okayama 700-8530, Japan
| | - Paul A Slesinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhenpeng Qin
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Steven O Nielsen
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
38
|
Basingab FS, Alshahrani OA, Alansari IH, Almarghalani NA, Alshelali NH, Alsaiary AH, Alharbi N, Zaher KA. From Pioneering Discoveries to Innovative Therapies: A Journey Through the History and Advancements of Nanoparticles in Breast Cancer Treatment. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:27-51. [PMID: 39867813 PMCID: PMC11761866 DOI: 10.2147/bctt.s501448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025]
Abstract
Nanoparticle technology has revolutionized breast cancer treatment by offering innovative solutions addressing the gaps in traditional treatment methods. This paper aimed to comprehensively explore the historical journey and advancements of nanoparticles in breast cancer treatment, highlighting their transformative impact on modern medicine. The discussion traces the evolution of nanoparticle-based therapies from their early conceptualization to their current applications and future potential. We initially explored the historical context of breast cancer treatment, highlighting the limitations of conventional therapies, such as surgery, radiation, and chemotherapy. The advent of nanotechnology has introduced a new era characterized by the development of various nanoparticles, including liposomes, dendrimers, and gold nanoparticles, designed to target cancer cells with remarkable precision. We further described the mechanisms of action for nanoparticles, including passive and active targeting, and reviewed significant breakthroughs and clinical trials that have validated their efficacy. Current applications of nanoparticles in breast cancer treatment have been examined, showcasing clinically approved therapies and comparing their effectiveness with traditional methods. This article also discusses the latest advancements in nanoparticle research, including drug delivery systems and combination therapy innovations, while addressing the current technical, biological, and regulatory challenges. The technical challenges include efficient and targeted delivery to tumor sites without affecting healthy tissue; biological, such as potential toxicity, immune system activation, or resistance mechanisms; economic, involving high production and scaling costs; and regulatory, requiring rigorous testing for safety, efficacy, and long-term effects to meet stringent approval standards. Finally, we have explored emerging trends, the potential for personalized medicine, and the ethical and social implications of this transformative technology. In conclusion, through comprehensive analysis and case studies, this paper underscores the profound impact of nanoparticles on breast cancer treatment and their future potential.
Collapse
Affiliation(s)
- Fatemah S Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Omniah A Alshahrani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Ibtehal H Alansari
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada A Almarghalani
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Nada H Alshelali
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Abeer Hamad Alsaiary
- Biology Department, College of Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Najwa Alharbi
- Department of Biology Science, Faculty of Science, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| | - Kawther A Zaher
- Immunology Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21859, Saudi Arabia
| |
Collapse
|
39
|
Zare D, Yılmaz G, Özçubukçu S. Application of Redox-Responsive Cysteine-Based Organogels as a Drug Delivery System for Doxorubicin. ACS OMEGA 2025; 10:147-156. [PMID: 39829527 PMCID: PMC11740374 DOI: 10.1021/acsomega.4c02620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 01/22/2025]
Abstract
Cysteine derivatives having disulfide bonds in their side chains can be used as redox-responsive organogelators. The disulfide bond can be cleaved in the presence of certain reducing agents like thiol derivatives such as glutathione (GSH), which is a tripeptide that consists of cysteine, glutamic acid, and glycine. Studies show that cells of certain cancers have higher levels of glutathione due to increased production of reactive oxygen species (ROS). This feature allows targeted cancer therapy using glutathione-responsive drug delivery systems. This study showed the drug delivery property of l-Cys(t-dodecyl-sulfanyl)-OH and l-Cys-(StBu)-OH-based organogels. The drug-release properties of these organogels were measured in the presence of GSH and were compared with the drug-release property of the l-Cys-(tBu)-OH-based organogel. The biocompatibility of the organogelators was measured in vitro by MTT assay and the characterization of microstructures and gel behaviors were studied using transmission electron microscopy (TEM) imaging, X-ray diffraction spectroscopy (XRD), Fourier-transform infrared spectroscopy (FTIR), and rheological measurements. The results indicated that the organogelators were able to form nanofibers by hydrogen bonds and van der Waals interactions between their hydrophobic groups and were able to release doxorubicin in the presence of GSH. The in vitro biocompatibility studies did not show significant toxicity to the L929 cells for l-Cys-(StBu)-OH and showed low concentrations of l-Cys(t-dodecyl-sulfanyl)-OH.
Collapse
Affiliation(s)
- Diba Zare
- Department of Chemistry, Middle East Technical University, 06800 Ankara, Turkey
| | - Gamze Yılmaz
- Department of Chemistry, Middle East Technical University, 06800 Ankara, Turkey
| | - Salih Özçubukçu
- Department of Chemistry, Middle East Technical University, 06800 Ankara, Turkey
| |
Collapse
|
40
|
Gleue L, Graefen B, Voigt M, Schupp J, Schneider D, Fichter M, Kuske M, Mailaender V, Tuettenberg A, Helm M. Dual Centrifugation-Based Screening for pH-Responsive Liposomes. ChemMedChem 2025; 20:e202400648. [PMID: 39328087 DOI: 10.1002/cmdc.202400648] [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: 08/19/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
In liposomal drug delivery development, the delicate balance of membrane stability is a major challenge to prevent leakage (during shelf-life and blood circulation), and to ensure efficient payload release at the therapeutic destination. Our composite screening approach uses the processing by dual centrifugation technique to speed up the identification of de novo formulations of intermediate membrane stability. By screening binary lipid combinations at systemically varied ratios we highlight liposomal formulations of intermediate stability, what we termed "the edge of stability", requiring moderate stimuli for destabilization. Supplementation with a pH-sensitive cholesterol derivative (to obtain acid labile liposomes) and renewed assessment with cargo load led to the discovery of three formulations with sufficient shelf-life stability, acceptable cargo retention and efficient pH-responsive cargo release in vitro. The "lead candidates" exhibited promising in cellulo uptake with increased intracellular cargo release and revealed in vivo performance advantages compared to a control liposome. Our approach filters lipid compositions on "the edge of stability" that were introduced with a pH-sensitive cholesterol derivate leading pH-responsive liposomes, out of a multidimensional parameter space. Their discovery by rational approaches would have been highly unlikely, thus highlighting the potential of our screening approach.
Collapse
Affiliation(s)
- Lukas Gleue
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, 55128, Mainz, Germany
| | - Barbara Graefen
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Matthias Voigt
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, 55128, Mainz, Germany
- BioNTech SE, An der Goldgrube 12, 55131, Mainz, Germany
| | - Jonathan Schupp
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Frankfurt Cancer Institute, Paul-Ehrlich-Straße 42-44, 60596, Frankfurt, Germany
- Goethe University Frankfurt, Institute of Neurology (Edinger Institute), Heinrich-Hoffmann-Straße 7, 60528, Frankfurt, Germany
| | - Dirk Schneider
- Department of Chemistry-Biochemistry, Johannes Gutenberg University, Hanns-Dieter-Hüsch-Weg 17, 55128, Mainz, Germany
| | - Michael Fichter
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Paul-Klein-Center for Immunintervention, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Michael Kuske
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Paul-Klein-Center for Immunintervention, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Institute for translational oncology Mainz (TRON-Mainz), Freiligrathstraße 12, 55131, Mainz, Germany
| | - Volker Mailaender
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Paul-Klein-Center for Immunintervention, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Mark Helm
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudingerweg 5, 55128, Mainz, Germany
| |
Collapse
|
41
|
Wahengbam GS, Nirmal S, Nandwana J, Kar S, Kumari V, Mishra R, Singh A. Polymeric Nanoparticles Revolutionizing Brain Cancer Therapy: A Comprehensive Review of Strategies and Advances. Crit Rev Ther Drug Carrier Syst 2025; 42:73-106. [PMID: 39819464 DOI: 10.1615/critrevtherdrugcarriersyst.2024051822] [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: 01/19/2025]
Abstract
Brain cancer continues to be one of the most formidable malignancies to manage, mainly attributable to the presence of the blood-brain barrier (BBB) limiting the permeability of drugs and the diverse characteristics of brain tumors complicating treatment. The management of brain tumors has been hampered by many different factors, including the impermeability of the BBB, which restricts the delivery of chemotherapeutic agents to the tumor site, as well as intertumoral heterogeneity and the influence of brain tumor stem cells. In addition, small molecular weight drugs cannot specifically accumulate in malignant cells and have a limited circulation half-life. Nanoparticles (NPs) can be engineered to traverse the BBB and transport therapeutic medications directly into the brain, enhancing their efficacy compared with the conventional delivery of unbound drugs. Surface modifications of NPs can boost their efficiency by increasing their selectivity towards tumor receptors. This review covers treatment methods for malignant gliomas, associated risk factors, and improvements in brain drug administration, emphasizing the future potential of polymeric NPs and their mechanism for crossing the BBB. To surmount these obstacles, the newly formulated drug-delivery approach utilizing NPs, particularly those coated with cell membranes, has demonstrated potential in treating brain cancer. These NPs provide targeted tumor specificity, biocompatibility, extended circulation, enhanced BBB penetration, and immune evasion. This review focuses on coating strategies for PLGA NPs, particularly dual-targeting methods, to enhance BBB permeability and tumor-targeted delivery of drugs in brain cancer.
Collapse
Affiliation(s)
| | - Sakshi Nirmal
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Jai Nandwana
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Swatileena Kar
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Vandana Kumari
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Rajeev Mishra
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, India
| | | |
Collapse
|
42
|
Juang V, Gan J, Xia Z, Wang Y, Schwendeman A. Development and optimization of an in vitro release assay for evaluation of liposomal irinotecan formulation. Int J Pharm 2024; 667:124854. [PMID: 39442767 DOI: 10.1016/j.ijpharm.2024.124854] [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: 08/22/2024] [Revised: 10/13/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Onivyde®, a pegylated irinotecan liposomal formulation, is approved by the USFDA for treating metastatic pancreatic adenocarcinoma. Despite the substantial interest in developing its generic versions, the unique structural and manufacturing complexities of liposomal formulations pose challenges. In this study, we address this gap by developing a robust in vitro release test (IVRT) using dialysis membrane techniques. The release of Onivyde® is influenced by several key factors, including the composition of the release medium, temperature, initial formulation concentration, the materials and molecular weight cut-offs of dialysis bags, and the pH of the release medium. Our optimized IVRT for Onivyde® incorporates a release medium containing 5 mM ammonium bicarbonate in a HEPES solution with a pH of 7.4. Additionally, the method includes an initial formulation concentration of 4.6 µg/mL and 50 kDa dialysis bags, while maintaining a temperature of 37 °C with continuous agitation at 80 rpm. This optimized IVR assay effectively differentiates between varying qualities of irinotecan liposomal formulations. Our findings contribute to optimizing IVRT for liposomal irinotecan formulations, enabling better quality control procedures. This assay serves as a reliable tool for evaluating generic irinotecan liposomal formulations, aiding in their development and ensuring in vitro comparability.
Collapse
Affiliation(s)
- Vivian Juang
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jingyao Gan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Ziyun Xia
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Yan Wang
- Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
43
|
Tsioulos G, Vallianou NG, Skourtis A, Dalamaga M, Kotsi E, Kargioti S, Adamidis N, Karampela I, Mourouzis I, Kounatidis D. Vaccination as a Promising Approach in Cardiovascular Risk Mitigation: Are We Ready to Embrace a Vaccine Strategy? Biomolecules 2024; 14:1637. [PMID: 39766344 PMCID: PMC11727084 DOI: 10.3390/biom14121637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular disease (CVD) remains a leading global health concern, with atherosclerosis being its principal cause. Standard CVD treatments primarily focus on mitigating cardiovascular (CV) risk factors through lifestyle changes and cholesterol-lowering therapies. As atherosclerosis is marked by chronic arterial inflammation, the innate and adaptive immune systems play vital roles in its progression, either exacerbating or alleviating disease development. This intricate interplay positions the immune system as a compelling therapeutic target. Consequently, immunomodulatory strategies have gained increasing attention, though none have yet reached widespread clinical adoption. Safety concerns, particularly the suppression of host immune defenses, remain a significant barrier to the clinical application of anti-inflammatory therapies. Recent decades have revealed the significant role of adaptive immune responses to plaque-associated autoantigens in atherogenesis, opening new perspectives for targeted immunological interventions. Preclinical models indicate that vaccines targeting specific atherosclerosis-related autoantigens can slow disease progression while preserving systemic immune function. In this context, numerous experimental studies have advanced the understanding of vaccine development by exploring diverse targeting pathways. Key strategies include passive immunization using naturally occurring immunoglobulin G (IgG) antibodies and active immunization targeting low-density lipoprotein cholesterol (LDL-C) and apolipoproteins, such as apolipoprotein B100 (ApoB100) and apolipoprotein CIII (ApoCIII). Other approaches involve vaccine formulations aimed at proteins that regulate lipoprotein metabolism, including proprotein convertase subtilisin/kexin type 9 (PCSK9), cholesteryl ester transfer protein (CETP), and angiopoietin-like protein 3 (ANGPTL3). Furthermore, the literature highlights the potential for developing non-lipid-related vaccines, with key targets including heat shock proteins (HSPs), interleukins (ILs), angiotensin III (Ang III), and a disintegrin and metalloproteinase with thrombospondin motifs 7 (ADAMTS-7). However, translating these promising findings into safe and effective clinical therapies presents substantial challenges. This review provides a critical evaluation of current anti-atherosclerotic vaccination strategies, examines their proposed mechanisms of action, and discusses key challenges that need to be overcome to enable clinical translation.
Collapse
Affiliation(s)
- Georgios Tsioulos
- Fourth Department of Internal Medicine, Medical School, Attikon General University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Alexandros Skourtis
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelia Kotsi
- Second Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Hippokratio General Hospital, 11527 Athens, Greece;
| | - Sofia Kargioti
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Nikolaos Adamidis
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Irene Karampela
- Second Department of Critical Care, Medical School, Attikon General University Hospital, University of Athens, 12461 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece;
| |
Collapse
|
44
|
Xiong L, Wei Y, Si H, Li Z, Wen J, Liu F, Wang X, Yang H, Chen L, Pi C, Han Y, Zhao L. Development of the Curcumin Analog CA7 Liposome and Its Evaluation for Efficacy Against Cervical Cancer in vitro and in vivo. Int J Nanomedicine 2024; 19:13411-13428. [PMID: 39691454 PMCID: PMC11651071 DOI: 10.2147/ijn.s493074] [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: 08/26/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Objective The objective of this study was to develop liposomes (LP) containing a curcumin (CU) analog CA7 to enhance its pharmacokinetic profile and anti-cervical cancer (CC) effects. Methods Single-factor and Box-Behnken experiments were conducted to optimize the formulation of CA7-loaded liposomes (CA7-LP). The in vitro release, stability, biocompatibility, and pharmacokinetic of CA7-LP were evaluated. The biological effects of CA7-LP on Hela cells were assessed using MTT assays, colony formation assays, wound healing assays, and flow cytometry. Additionally, the anti-CC efficacy of CA7-LP was tested in mouse models of transplanted tumors. Results The optimal formulation of CA7-LP exhibited a particle size of 92.43 ± 1.52 nm, a polydispersity index of 0.27 ± 0.01, an encapsulation efficiency of 97.79 ± 1.49%, a drug loading of 3.23 ± 0.20%, and a zeta potential of -6.69 ± 0.77 mV. Transmission electron microscopy confirmed that a spherical morphology was exhibited by CA7-LP. The cumulative in vitro release of CA7-LP was found to be 2.84 times greater than that of CA7, and stability at room temperature was maintained for at least 90 d. Furthermore, a significantly higher uptake of CA7-LP by Hela cells was observed compared to curcumin and CA7, leading to enhanced inhibition of cell proliferation, migration and cell cycle, as well as increased apoptosis (p < 0.05). In vivo studies revealed that CA7-LP exhibited superior pharmacokinetic properties compared to CA7 (AUC: 3.58-fold, Cmax: 5.65-fold, t1/2z: 1.2-fold). The anti-CC effects of CA7-LP were found to be comparable to those of Cisplatin injection, with a better safety profile. Conclusion The newly developed CA7-LP is considered a promising candidate for the treatment of CC, demonstrating high potential for clinical application.
Collapse
Affiliation(s)
- Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Hui Si
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Zheng Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Jie Wen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Furong Liu
- Department of Oncology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Xiaodong Wang
- Department of Hepatobiliary Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Hongru Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Ling Zhao
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| |
Collapse
|
45
|
Tomsen-Melero J, Moltó-Abad M, Merlo-Mas J, Díaz-Riascos ZV, Cristóbal-Lecina E, Soldevila A, Altendorfer-Kroath T, Danino D, Ionita I, Pedersen JS, Snelling L, Clay H, Carreño A, Corchero JL, Pulido D, Casas J, Veciana J, Schwartz S, Sala S, Font A, Birngruber T, Royo M, Córdoba A, Ventosa N, Abasolo I, González-Mira E. Targeted nanoliposomes to improve enzyme replacement therapy of Fabry disease. SCIENCE ADVANCES 2024; 10:eadq4738. [PMID: 39671483 PMCID: PMC11801267 DOI: 10.1126/sciadv.adq4738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024]
Abstract
The central nervous system represents a major target tissue for therapeutic approach of numerous lysosomal storage disorders. Fabry disease arises from the lack or dysfunction of the lysosomal alpha-galactosidase A (GLA) enzyme, resulting in substrate accumulation and multisystemic clinical manifestations. Current enzyme replacement therapies (ERTs) face limited effectiveness due to poor enzyme biodistribution in target tissues and inability to reach the brain. We present an innovative drug delivery strategy centered on a peptide-targeted nanoliposomal formulation, designated as nanoGLA, engineered to selectively deliver a recombinant human GLA (rhGLA) to target tissues. In a Fabry mouse model, nanoGLA demonstrated improved efficacy, inducing a notable reduction in Gb3 deposits in contrast to non-nanoformulated GLA, even in the brain, highlighting the potential of the nanoGLA to address both systemic and cerebrovascular manifestations of Fabry disease. The EMA has granted the Orphan Drug Designation to this product, underscoring the potential clinical superiority of nanoGLA over authorized ERTs and encouraging to advance it toward clinical translation.
Collapse
Affiliation(s)
- Judit Tomsen-Melero
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - Marc Moltó-Abad
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Josep Merlo-Mas
- Nanomol Technologies SL, Campus de la UAB, 08193 Bellaterra, Spain
| | - Zamira V. Díaz-Riascos
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Functional Validaton & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Edgar Cristóbal-Lecina
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Química Avançada de Catalunya (IQAC-CSIC), c/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | | | - Thomas Altendorfer-Kroath
- JOANNEUM RESEARCH–Institute for Biomedical Research and Technologies (HEALTH), Neue Stiftingtalstraße 2, 8010 Graz, Austria
| | - Dganit Danino
- Cryo-EM Laboratory of Soft Matter, Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, 32000 Haifa, Israel
- Cryo-EM and Self-Assembly Laboratory, Guangdong-Technion Israel Institute of Technology, Shantou, China
| | - Inbal Ionita
- Cryo-EM Laboratory of Soft Matter, Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Jan Skov Pedersen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus C, Denmark
| | | | - Hazel Clay
- Labcorp Drug Development, Harrogate HG3 IPY, UK
| | - Aida Carreño
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - José L. Corchero
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i de Microbiologia, Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Daniel Pulido
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Química Avançada de Catalunya (IQAC-CSIC), c/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Josefina Casas
- Institut de Química Avançada de Catalunya (IQAC-CSIC), c/ Jordi Girona 18-26, 08034 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain
| | - Jaume Veciana
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - Simó Schwartz
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Servei de Bioquímica, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Santi Sala
- Nanomol Technologies SL, Campus de la UAB, 08193 Bellaterra, Spain
| | | | - Thomas Birngruber
- JOANNEUM RESEARCH–Institute for Biomedical Research and Technologies (HEALTH), Neue Stiftingtalstraße 2, 8010 Graz, Austria
| | - Miriam Royo
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Química Avançada de Catalunya (IQAC-CSIC), c/ Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Alba Córdoba
- Nanomol Technologies SL, Campus de la UAB, 08193 Bellaterra, Spain
| | - Nora Ventosa
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - Ibane Abasolo
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Biochemistry, Drug Delivery & Targeting (CB-DDT), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Functional Validaton & Preclinical Research (FVPR)/U20 ICTS Nanbiosis, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Servei de Bioquímica, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Elisabet González-Mira
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| |
Collapse
|
46
|
Fernández-García R, Fraguas-Sánchez AI. Nanomedicines for Pulmonary Drug Delivery: Overcoming Barriers in the Treatment of Respiratory Infections and Lung Cancer. Pharmaceutics 2024; 16:1584. [PMID: 39771562 PMCID: PMC11677881 DOI: 10.3390/pharmaceutics16121584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The pulmonary route for drug administration has garnered a great deal of attention in therapeutics for treating respiratory disorders. It allows for the delivery of drugs directly to the lungs and, consequently, the maintenance of high concentrations at the action site and a reduction in systemic adverse effects compared to other routes, such as oral or intravenous. Nevertheless, the pulmonary administration of drugs is challenging, as the respiratory system tries to eliminate inhaled particles, being the main responsible mucociliary escalator. Nanomedicines represent a primary strategy to overcome the limitations of this route as they can be engineered to prolong pulmonary retention and avoid their clearance while reducing drug systemic distribution and, consequently, systemic adverse effects. This review analyses the use of pulmonary-administered nanomedicines to treat infectious diseases affecting the respiratory system and lung carcinoma, two pathologies that represent major health threats.
Collapse
Affiliation(s)
| | - Ana I. Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Complutense University, 28040 Madrid, Spain
- Institute of Industrial Pharmacy, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
47
|
Siriwardane DA, Shakiba S, Jiang W, Mudalige T. Evaluation of size-based distribution of components in VYXEOS® liposomal formulation using asymmetric flow field-flow fractionation. J Chromatogr A 2024; 1738:465488. [PMID: 39515205 DOI: 10.1016/j.chroma.2024.465488] [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: 07/23/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
VYXEOS® is the first FDA-approved dual-API liposomal formulation containing two different chemotherapeutics, daunorubicin and cytarabine at a 1:5 molar ratio. Analysis of bulk formulation does not provide insight to size-based distribution of APIs and excipients, therefore asymmetrical flow field-flow fractionation (AF4) was utilized for the size-based separation of VYXEOS® liposomes and collected size fractions were further analyzed for the concentrations of APIs, lipid excipients, and copper. Analysis results revealed a significant variation in API distribution across the size fractions, with the larger liposomes encapsulating a higher ratio of cytarabine to daunorubicin compared to the smaller liposomes, while lipid excipient composition was held constant across the size range. We attribute the size-based variation of API ratios to structural change during API loading and sequence of API loading. Cytarabine is first passively loaded into liposomes containing copper gluconate, and then daunorubicin is actively loaded using copper gradient where the daunorubicin is retained in the liposomes as a copper complex. This method can be extended to characterize various single and dual-API liposomal nanocarriers during drug product development and/or post market quality control.
Collapse
Affiliation(s)
- Dumindika A Siriwardane
- Arkansas Laboratory, Office of Regulatory Science, Office of Regulatory Affairs, U.S. Food and Drug Administration, Jefferson, AR 72079, United States
| | - Sheyda Shakiba
- Arkansas Laboratory, Office of Regulatory Science, Office of Regulatory Affairs, U.S. Food and Drug Administration, Jefferson, AR 72079, United States
| | - Wenlei Jiang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food, and Drug Administration, Silver Spring, MD 20993, United States.
| | - Thilak Mudalige
- Arkansas Laboratory, Office of Regulatory Science, Office of Regulatory Affairs, U.S. Food and Drug Administration, Jefferson, AR 72079, United States.
| |
Collapse
|
48
|
Yuan M, Wang Y, Wan Y, Li S, Tang J, Liang X, Zeng B, Li M, Wei X, Li X, Guo L, Guo Y. Novel sodium tauroursodeoxycholate-based multifunctional liposomal delivery system for encapsulation of oleanolic acid and combination therapy of type 2 diabetes mellitus. Int J Pharm 2024; 666:124803. [PMID: 39368671 DOI: 10.1016/j.ijpharm.2024.124803] [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: 07/04/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Liposomes have demonstrated great potential for drug delivery and diabetes treatment. However, hydrolysis by enzymes and emulsification by endogenous bile salts make liposomes unstable in the gastrointestinal tract. In this study, sodium tauroursodeoxycholate (TUDCNa)-based multifunctional bilosomes were designed to address the deficiencies of conventional liposomes. In the designed bilosomes, cholesterol was replaced by TUDCNa, which served as both a membrane stabilizer and an antidiabetic drug. Oleanolic acid (OA) was encapsulated in both conventional liposomes (OA-Ch-Lip) and bilosomes (OA-Tu-Bil) to compare their properties. Firstly, OA-Tu-Bil exhibited similar encapsulation efficiency and drug loading compared to OA-Ch-Lip, but with a smaller particle size. Secondly, OA-Tu-Bil showed better stability than OA-Ch-Lip. Thirdly, bilosomes exhibited prolonged intestinal retention time and improved permeability and oral bioavailability. Fourthly, in type 2 diabetes mellitus (T2DM) mice model, TUDCNa synergized with OA to exhibit the strongest therapeutic effect. In conclusion, TUDCNa have demonstrated the ability to substitute cholesterol in conventional liposomes, it provided a new approach for oral delivery of hypoglycemic drugs, and offered an innovative strategy for combination therapy.
Collapse
Affiliation(s)
- Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Xue Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Bin Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Meifeng Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Xiaohang Wei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Xiaohong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| |
Collapse
|
49
|
Bujan A, del Valle Alonso S, Chiaramoni NS. Photopolymerizable robust lipids towards reliability and their applications. Biophys Rev 2024; 16:773-782. [PMID: 39830130 PMCID: PMC11735817 DOI: 10.1007/s12551-024-01221-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/22/2024] [Indexed: 01/22/2025] Open
Abstract
Synthetic lipids have been studied as components in membrane models and drug delivery systems. Polymerizable phospholipids, especially photosensitive ones, can form new bilayer bonds when UV light irradiates. These phospholipids have been known since the 1980s, but in the last few years, new applications have been highlighted. Its use in drug delivery systems is interesting since the photopolymerization reaction produces highly stable vesicles. Additionally, the rearrangement of the acyl chains during the photopolymerization process can be applied in the generation of pores, resulting in systems that serve for drug-controlled release. In this article, our goal was to envision earlier photopolymers' publications towards the implications of these versatile phospholipids that led to proposed systems for drug delivery and controlled release of drugs at specific sites. This review offers a broad background towards a simple, reliable, and robust platform to make its application available.
Collapse
Affiliation(s)
- Ariana Bujan
- Laboratorio de BioNanotecnología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- CONICET - Facultad de Ciencias Exactas, Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Silvia del Valle Alonso
- Laboratorio de BioNanotecnología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- Grupo de Biología Estructural y Biotecnología (GBEyB), IMBICE (CONICET CCT-La Plata), Buenos Aires, Argentina
| | - Nadia S. Chiaramoni
- Laboratorio de BioNanotecnología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Buenos Aires, Argentina
- Grupo de Biología Estructural y Biotecnología (GBEyB), IMBICE (CONICET CCT-La Plata), Buenos Aires, Argentina
| |
Collapse
|
50
|
Abawi A, Trunfio-Sfarghiu AM, Thomann C, Petiot E, Lollo G, Granjon T, Girard-Egrot A, Maniti O. Tailor-made vincristine-liposomes for tumor targeting. Biochimie 2024; 227:35-46. [PMID: 39094823 DOI: 10.1016/j.biochi.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
To ensure selective targeting based on membrane fluidity and physico-chemical compatibility between the biological membrane of the target cell and the lipid membrane of the liposomes carriers. Lipid-based carriers as liposomes with varying membrane fluidities were designed for delivering vincristine, an anti-tumor compound derived from Madagascar's periwinkle. Liposomes, loaded with vincristine, were tested on prostate, colon, and breast cancer cell lines alongside non-tumor controls. Results showed that vincristine-loaded liposomes with fluid membranes significantly decreased the viability of cancer cell lines compared to controls. Confocal microscopy revealed the intracellular release of vincristine, evidenced by disrupted mitosis-specific labeling of actin filaments in metastatic prostate cell lines. This highlights the crucial role of membrane fluidity in the development of lipid-based drug carriers, offering a promising and cost-effective option for targeting cancer cells as an alternative to conventional strategies.
Collapse
Affiliation(s)
- Ariana Abawi
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | | | - Céline Thomann
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Emma Petiot
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Giovanna Lollo
- Laboratoire D'Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Thierry Granjon
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Agnès Girard-Egrot
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Ofelia Maniti
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| |
Collapse
|