1
|
Tuna RW, Achmad NA, Kurniawan I, Khairiyah, Asaf MB, Sapiun Z, Himawan A, Dominguez-Robles J, Aswad M, Permana AD. Development of thermosensitive mucoadhesive gel incorporated lipid microspheres of donepezil for enhanced nose-to-brain delivery. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-28. [PMID: 40262568 DOI: 10.1080/09205063.2025.2492455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
Alzheimer's disease (ALZ) is a chronic disease that affects the brain neurons leading to dementia. Donepezil (DPZ), a first-line treatment for ALZ is a potent symptomatic therapeutic agent. However, the oral and transdermal route represents non-targeted delivery, causing various adverse effects. This study presents the successful incorporation of a DPZ-loaded lipid microsphere (DPZ-LM) system into a thermosensitive-mucoadhesive gel (TMG), thereby enhancing the delivery of DPZ through the nose-to-brain route. To optimize the formulations, several evaluations were conducted, resulting in an optimized formulation of LM using Compritol® exhibited particle size of 8.75 µm, 98.44% of DPZ entrapped, and 93.40% of DPZ loaded in the system with a sustained release manner in the in vitro studies. The TMG-DPZ-LM was prepared using Pluronic® F127 and F68, as the gelling agents, with the addition of sodium alginate, as the mucoadhesive polymer. Following incorporation into TMG-DPZ-LM, the system exhibited excellent physicochemical properties and effective nasal delivery in ex vivo permeation has found that 88.58 ± 12.53 µg/cm2 and retention studies with a mean concentration of 0.0077 mg of retention DPZ in porcine nasal mucosa. The in vivo pharmacokinetic studies demonstrated that the administration of TMG-DPZ-LM via the nose-to-brain route resulted in a significant (p < 0.05) increase in the Cmax, with 207.24 ± 5.16 µg/cm3 of DPZ in the brain that exhibited a significantly different profile compared to the other route and formulation. The TMG-DPZ-LM system that was developed in this study was considered to have improved its efficacy in the treatment of ALZ.
Collapse
Affiliation(s)
- Rachmatya W Tuna
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Nurafni Annisa Achmad
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Irfan Kurniawan
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Khairiyah
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Muh Bisfain Asaf
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Zulfiayu Sapiun
- Department of Pharmacy, Health Polytechnic of Gorontalo, Gorontalo, Indonesia
| | - Achmad Himawan
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Juan Dominguez-Robles
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
| | - Muhammad Aswad
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Andi Dian Permana
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
2
|
Azam U, Naseer MM, Rochais C. Analysis of skeletal diversity of multi-target directed ligands (MTDLs) targeting Alzheimer's disease. Eur J Med Chem 2025; 286:117277. [PMID: 39848035 DOI: 10.1016/j.ejmech.2025.117277] [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: 10/11/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/25/2025]
Abstract
Alzheimer's disease (AD) remains a significant healthcare challenge, necessitating innovative therapeutic approaches to address its complex and multifactorial nature. Traditional drug discovery strategies targeting single molecular targets are not sufficient for the effective treatment of AD. In recent years, MTDLs have emerged as promising candidates for AD therapy, aiming to simultaneously modulate multiple pathological targets. Among the various strategies employed in MTDL design, pharmacophore hybridization offers a versatile approach to integrate diverse pharmacophoric features within a single molecular scaffold. This strategy provides access to a wide array of chemical space for the design and development of novel therapeutic agents. This review, therefore, provides a comprehensive overview of skeletal diversity exhibited by MTDLs designed recently for AD therapy based on pharmacophore hybridization approach. A diverse range of pharmacophoric elements and core scaffolds hybridized to construct MTDLs that has the potential to target multiple pathological features of AD including amyloid-beta aggregation, tau protein hyperphosphorylation, cholinergic dysfunction, oxidative stress, and neuroinflammation are discussed. Through the comprehensive analysis and integration of structural insights of key biomolecular targets, this review aims to enhance optimization efforts in MTDL design, ultimately striving towards a comprehensive cure for the multifaceted pathophysiology of the disease.
Collapse
Affiliation(s)
- Uzma Azam
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Moazzam Naseer
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan; Université de Caen Normandie, Normandie Univ., CERMN, 14000, Caen, France.
| | - Christophe Rochais
- Université de Caen Normandie, Normandie Univ., CERMN, 14000, Caen, France.
| |
Collapse
|
3
|
Guler E, Yekeler HB, Uner B, Dogan M, Asghar A, Ikram F, Yazir Y, Gunduz O, Kalaskar DM, Cam ME. In Vitro Neuroprotective Effect Evaluation of Donepezil‐Loaded PLGA Nanoparticles‐Embedded PVA/PEG Nanofibers on SH‐SY5Y Cells and AP‐APP Plasmid Related Alzheimer Cell Line Model. MACROMOLECULAR MATERIALS AND ENGINEERING 2025; 310. [DOI: 10.1002/mame.202400160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Indexed: 03/30/2025]
Abstract
AbstractRecently developed nanoparticles and nanofibers present new brain‐specific treatment strategies, especially for Alzheimer's disease treatment. In this study, donepezil (DO)‐loaded PLGA nanoparticles (DNP) are embedded in PVA/PEG nanofibers (DNPF) produced by pressurized gyration for sublingual administration. SEM images showed produced drug‐loaded and pure nanofibers, which have sizes between 978 and 1123 nm, demonstrated beadless morphology and homogeneous distribution. FT‐IR, XRD, and DSC results proved the produced nanoparticles and fibers to consist of the DO and other polymers. The in vitro drug release test presented that the release profile of DO is completed at the end of the 18th day. It is released by the first order kinetic model. DNPF has an ultra‐fast release profile via its disintegration within 2 sec, which proved itself to be suitable for the administration sublingually. All samples presented above ≈90% cell viability via their non‐toxic natures on SH‐SY5Y human neuroblastoma cells by using Alamar blue assay. The anti‐Alzheimer effects of DO, DNP, and DNPF are evaluated on the Aβ1−42‐induced SH‐SY5Y cells at 1, 5, and 10 µM as treatment groups. The 1 µM dosage exhibited the most significant neuroprotective effects, which showed enhanced cellular uptake and superior modulation of Alzheimer's‐related proteins, including tau and Aβ.
Collapse
Affiliation(s)
- Ece Guler
- Department of Pharmacology, School of Pharmacy Istanbul Kent University Istanbul 34406 Kagithane Türkiye
- Center for Nanotechnology and Biomaterials Application and Research Marmara University Istanbul 34722 Türkiye
- UCL Division of Surgery and Interventional Sciences Rowland Hill Street London NW3 2PF UK
- MecNano Technologies Cube Incubation Istanbul 34906 Teknopark İstanbul Türkiye
| | - Humeyra Betul Yekeler
- Center for Nanotechnology and Biomaterials Application and Research Marmara University Istanbul 34722 Türkiye
- UCL Division of Surgery and Interventional Sciences Rowland Hill Street London NW3 2PF UK
- MecNano Technologies Cube Incubation Istanbul 34906 Teknopark İstanbul Türkiye
- Department of Pharmacology, Faculty of Pharmacy Marmara University Istanbul 34854 Türkiye
| | - Burcu Uner
- MecNano Technologies Cube Incubation Istanbul 34906 Teknopark İstanbul Türkiye
- Department of Pharmaceutical Technology, School of Pharmacy Istanbul Kent University Istanbul 34406 Türkiye
- Department of Pharmaceutical and Administrative Science University of Health Science and Pharmacy in St. Louis St. Louis 63110 MO USA
- Department of Anesthesiology Center for Clinical Pharmacology Washington University School of Medicine in St. Louis St. Louis 63110 MO USA
| | - Murat Dogan
- Department of Pharmaceutical Biotechnology Faculty of Pharmacy Sivas Cumhuriyet University Sivas 58140 Türkiye
- Cancer Survivorship Institute Robert H. Lurie Comprehensive Cancer Center Northwestern University 625 N. Michigan Ave., Suite 2100 Chicago 60611 IL USA
| | - Asima Asghar
- Interdisciplinary Research Centre in Biomedical Materials COMSATS University Islamabad Lahore Campus Pakistan
| | - Fakhera Ikram
- Interdisciplinary Research Centre in Biomedical Materials COMSATS University Islamabad Lahore Campus Pakistan
| | - Yusufhan Yazir
- Stem Cell and Gene Therapies Research and Applied Center, Medical Faculty Kocaeli University Kocaeli 41380 Turkiye
| | - Oguzhan Gunduz
- Center for Nanotechnology and Biomaterials Application and Research Marmara University Istanbul 34722 Türkiye
- Department of Metallurgical and Materials Engineering Faculty of Technology Marmara University Istanbul 34730 Türkiye
| | - Deepak M Kalaskar
- UCL Division of Surgery and Interventional Sciences Rowland Hill Street London NW3 2PF UK
| | - Muhammet Emin Cam
- Department of Pharmacology, School of Pharmacy Istanbul Kent University Istanbul 34406 Kagithane Türkiye
- UCL Division of Surgery and Interventional Sciences Rowland Hill Street London NW3 2PF UK
- MecNano Technologies Cube Incubation Istanbul 34906 Teknopark İstanbul Türkiye
- Biomedical Engineering Department University of Aveiro Aveiro 3810‐193 Portugal
| |
Collapse
|
4
|
Guler E, Yekeler HB, Uner B, Dogan M, Asghar A, Ikram F, Yazir Y, Gunduz O, Kalaskar DM, Cam ME. In Vitro Neuroprotective Effect Evaluation of Donepezil‐Loaded PLGA Nanoparticles‐Embedded PVA/PEG Nanofibers on SH‐SY5Y Cells and AP‐APP Plasmid Related Alzheimer Cell Line Model. MACROMOLECULAR MATERIALS AND ENGINEERING 2025; 310. [DOI: 4.https:/doi.org/10.1002/mame.202400160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Indexed: 03/30/2025]
Abstract
AbstractRecently developed nanoparticles and nanofibers present new brain‐specific treatment strategies, especially for Alzheimer's disease treatment. In this study, donepezil (DO)‐loaded PLGA nanoparticles (DNP) are embedded in PVA/PEG nanofibers (DNPF) produced by pressurized gyration for sublingual administration. SEM images showed produced drug‐loaded and pure nanofibers, which have sizes between 978 and 1123 nm, demonstrated beadless morphology and homogeneous distribution. FT‐IR, XRD, and DSC results proved the produced nanoparticles and fibers to consist of the DO and other polymers. The in vitro drug release test presented that the release profile of DO is completed at the end of the 18th day. It is released by the first order kinetic model. DNPF has an ultra‐fast release profile via its disintegration within 2 sec, which proved itself to be suitable for the administration sublingually. All samples presented above ≈90% cell viability via their non‐toxic natures on SH‐SY5Y human neuroblastoma cells by using Alamar blue assay. The anti‐Alzheimer effects of DO, DNP, and DNPF are evaluated on the Aβ1−42‐induced SH‐SY5Y cells at 1, 5, and 10 µM as treatment groups. The 1 µM dosage exhibited the most significant neuroprotective effects, which showed enhanced cellular uptake and superior modulation of Alzheimer's‐related proteins, including tau and Aβ.
Collapse
Affiliation(s)
- Ece Guler
- Department of Pharmacology, School of Pharmacy Istanbul Kent University Istanbul 34406 Kagithane Türkiye
- Center for Nanotechnology and Biomaterials Application and Research Marmara University Istanbul 34722 Türkiye
- UCL Division of Surgery and Interventional Sciences Rowland Hill Street London NW3 2PF UK
- MecNano Technologies Cube Incubation Istanbul 34906 Teknopark İstanbul Türkiye
| | - Humeyra Betul Yekeler
- Center for Nanotechnology and Biomaterials Application and Research Marmara University Istanbul 34722 Türkiye
- UCL Division of Surgery and Interventional Sciences Rowland Hill Street London NW3 2PF UK
- MecNano Technologies Cube Incubation Istanbul 34906 Teknopark İstanbul Türkiye
- Department of Pharmacology, Faculty of Pharmacy Marmara University Istanbul 34854 Türkiye
| | - Burcu Uner
- MecNano Technologies Cube Incubation Istanbul 34906 Teknopark İstanbul Türkiye
- Department of Pharmaceutical Technology, School of Pharmacy Istanbul Kent University Istanbul 34406 Türkiye
- Department of Pharmaceutical and Administrative Science University of Health Science and Pharmacy in St. Louis St. Louis 63110 MO USA
- Department of Anesthesiology Center for Clinical Pharmacology Washington University School of Medicine in St. Louis St. Louis 63110 MO USA
| | - Murat Dogan
- Department of Pharmaceutical Biotechnology Faculty of Pharmacy Sivas Cumhuriyet University Sivas 58140 Türkiye
- Cancer Survivorship Institute Robert H. Lurie Comprehensive Cancer Center Northwestern University 625 N. Michigan Ave., Suite 2100 Chicago 60611 IL USA
| | - Asima Asghar
- Interdisciplinary Research Centre in Biomedical Materials COMSATS University Islamabad Lahore Campus Pakistan
| | - Fakhera Ikram
- Interdisciplinary Research Centre in Biomedical Materials COMSATS University Islamabad Lahore Campus Pakistan
| | - Yusufhan Yazir
- Stem Cell and Gene Therapies Research and Applied Center, Medical Faculty Kocaeli University Kocaeli 41380 Turkiye
| | - Oguzhan Gunduz
- Center for Nanotechnology and Biomaterials Application and Research Marmara University Istanbul 34722 Türkiye
- Department of Metallurgical and Materials Engineering Faculty of Technology Marmara University Istanbul 34730 Türkiye
| | - Deepak M Kalaskar
- UCL Division of Surgery and Interventional Sciences Rowland Hill Street London NW3 2PF UK
| | - Muhammet Emin Cam
- Department of Pharmacology, School of Pharmacy Istanbul Kent University Istanbul 34406 Kagithane Türkiye
- UCL Division of Surgery and Interventional Sciences Rowland Hill Street London NW3 2PF UK
- MecNano Technologies Cube Incubation Istanbul 34906 Teknopark İstanbul Türkiye
- Biomedical Engineering Department University of Aveiro Aveiro 3810‐193 Portugal
| |
Collapse
|
5
|
Pei J, Kumarasamy RV, Jayaraman S, Kanniappan GV, Long Q, Palanisamy CP. Quercetin-functionalized nanomaterials: Innovative therapeutic avenues for Alzheimer's disease management. Ageing Res Rev 2025; 104:102665. [PMID: 39824363 DOI: 10.1016/j.arr.2025.102665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/20/2025]
Abstract
Alzheimer's Disease (AD) is a major global health challenge, largely due to its complex pathology and the limited effectiveness of existing treatments. Quercetin, a bioactive compound belonging to the flavonoid class, its promising antioxidant, anti-inflammatory, and neuroprotective effects in addressing AD. However, its therapeutic potential is hindered by challenges such as low bioavailability, instability, and restricted permeability across the blood-brain barrier (BBB). Advances in nanotechnology have paved the way for quercetin-functionalized nanomaterials, offering solutions to these challenges. These nanostructures enhance quercetin's solubility, stability, and targeted brain delivery, thereby augmenting its therapeutic potential. In this review, nanocarriers (like liposomes, polymeric nanoparticles, and metal-based nanosystems) are explored for their potential application in optimizing quercetin delivery in AD management. It discusses the mechanisms by which these nanostructures enhance BBB penetration and exert neuroprotective effects. Furthermore, the review examines the outcomes of preclinical and in vitro studies, while addressing the challenges of scaling these approaches for clinical application. By merging the fields of nanotechnology and neurotherapeutics, the importance of quercetin-functionalized nanomaterials in advancing AD management strategies is underscored in this review.
Collapse
Affiliation(s)
- Jinjin Pei
- College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Pro-cessing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guang-dong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Guangdong Ocean University, Zhanjiang 524088, China
| | | | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Gopalakrishnan Velliyur Kanniappan
- Department of physiology, Saveetha Medical College & Hospital (SMCH), Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu 602105, India.
| | - Qianfa Long
- Department of Neurosurgery, Xi'an Central Hospital, Xi'an Jiaotong University, No. 161, West 5th Road, Xincheng District, Xi'an 710003, PR China.
| | - Chella Perumal Palanisamy
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
6
|
Okafor NI, Omoteso OA, Choonara YE. The modification of conventional liposomes for targeted antimicrobial delivery to treat infectious diseases. DISCOVER NANO 2025; 20:19. [PMID: 39883380 PMCID: PMC11782757 DOI: 10.1186/s11671-024-04170-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
Some of the most crucial turning points in the treatment strategies for some major infectious diseases including AIDS, malaria, and TB, have been reached with the introduction of antimicrobials and vaccines. Drug resistance and poor effectiveness are key limitations that need to be overcome. Conventional liposomes have been explored as a delivery system for infectious diseases bioactives to treat infectious diseases to provide an efficient approach to maximize the therapeutic outcomes, drug stability, targetability, to reduce the side-effects of antimicrobials, and enhance vaccine performance where necessary. However, as the pathological understanding of infectious diseases become more known, the need for more advanced liposomal technologies was born to continue having a profound effect on targeted chemotherapy for infectious diseases. This review therefore provides a concise incursion into the most recent and vogue liposomal formulations used to treat infectious diseases. An appraisal of immunological, stimuli-responsive, biomimetic and functionalized liposomes and other novel modifications to conventional liposomes is assimilated in sync with mutations of resistant pathogens.
Collapse
Affiliation(s)
- Nnamdi Ikemefuna Okafor
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | | | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
7
|
Achmad NA, Tuna RW, Kurniawan I, Khairiyah, Asaf MB, Rahman L, Manggau MA, Aliyah, Dominguez-Robles J, Aswad M, Permana AD. Development of Thermosensitive Mucoadhesive Gel Based Encapsulated Lipid Microspheres as Nose-to-Brain Rivastigmine Delivery System. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:314-328. [PMID: 39714110 DOI: 10.1021/acs.langmuir.4c03530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Alzheimer's disease (ALZ) is a neurodegenerative disease that damages neuronal cells and causes decline in cognitive abilities. Administration of cholinesterase inhibitor compounds is the primary choice in the treatment of ALZ, one of which is rivastigmine (RVT). Several routes of administration of RVT are available, such as oral and transdermal. However, in the oral route, RVT has low bioavailability, undergoes first-pass metabolism, and the presence of the blood-brain barrier (BBB) reduces the therapeutic concentration of RVT. The transdermal route is nonselective target in the brain. This study aims to combine thermosensitive mucoadhesive gel (TG) and lipid microspheres (LM) as a drug delivery system to improve the efficacy of RVT. Combining these will prevent systemic side effects of RVT and increase drug concentration in the brain. LM was formulated with varying concentrations of Compritol polymer. The results of LM evaluation showed the values of particle size, PDI, and %EE and %DL were 8.519 μm, 0.018 ± 0.004, 72.54%, and 76.43%, respectively. The TG formulation can provide a liquid form at room temperature (25 °C) and a gel at nasal temperature (35 °C). Hemolytic and HET-CAM tests confirmed TG RVT LM's safety for use. Ex vivo studies showed controlled and sustained release of TG RVT LM, and in vivo studies showed TG RVT LM a higher pharmacokinetic profile in the brain than oral formulations and injections. The Cmax was found to be 7.05 ± 0.55 μg/cm3, Tmax was 24 h, and AUC0-24, which is related to the effectiveness of brain targeting, was 225.73 μg/cm3. In conclusion, this study shows the successful development of TG RVT LM, as evidenced by improved drug delivery to the brain, which is characterized by higher concentrations of RVT in the brain compared with oral and injectable RVT, this delivery system shows potential as a future treatment for Alzheimer's disease.
Collapse
Affiliation(s)
- Nurafni Annisa Achmad
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Rachmatya W Tuna
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Irfan Kurniawan
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Khairiyah
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Muhammad Bisfain Asaf
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Latifah Rahman
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Marianti A Manggau
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Aliyah
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Juan Dominguez-Robles
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Seville 41012, Spain
| | - Muhammad Aswad
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| | - Andi Dian Permana
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi, Indonesia
| |
Collapse
|
8
|
Inamdar A, Gurupadayya B, Halagali P, Nandakumar S, Pathak R, Singh H, Sharma H. Cutting-edge Strategies for Overcoming Therapeutic Barriers in Alzheimer's Disease. Curr Pharm Des 2025; 31:598-618. [PMID: 39492772 DOI: 10.2174/0113816128344571241018154506] [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: 08/04/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) remains one of the hardest neurodegenerative diseases to treat due to its enduring cognitive deterioration and memory loss. Despite extensive research, few viable treatment approaches have been found; these are mostly due to several barriers, such as the disease's complex biology, limited pharmaceutical efficacy, and the BBB. This presentation discusses current strategies for addressing these therapeutic barriers to enhance AD treatment. Innovative drug delivery methods including liposomes, exosomes, and nanoparticles may be able to pass the blood-brain barrier and allow medicine to enter specific brain regions. These innovative strategies of medicine distribution reduce systemic side effects by improving absorption. Moreover, the development of disease-modifying treatments that target tau protein tangles, amyloid-beta plaques, and neuroinflammation offers the chance to influence the course of the illness rather than only treat its symptoms. Furthermore, gene therapy and CRISPR-Cas9 technologies have surfaced as potentially groundbreaking methods for addressing the underlying genetic defects associated with AD. Furthermore, novel approaches to patient care may involve the utilization of existing medications having neuroprotective properties, such as those for diabetes and cardiovascular conditions. Furthermore, biomarker research and personalized medicine have made individualized therapy approaches possible, ensuring that patients receive the best care possible based on their unique genetic and molecular profiles.
Collapse
Affiliation(s)
- Aparna Inamdar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Bannimath Gurupadayya
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Prashant Halagali
- Department of Pharmaceutical Quality Assurance, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi 590010, Karnataka, India
| | - S Nandakumar
- Associate Scientist, Corteva Agriscience, Hyderabad 500081, Telangana, India
| | - Rashmi Pathak
- Department of Pharmacy, Invertis University, Bareilly (UP) 243123, India
| | - Himalaya Singh
- Department of Medicine, Government Institute of Medical Sciences, Greater Noida (UP) 201312, India
| | - Himanshu Sharma
- Department of Pharmacy, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad (UP) 244001, India
| |
Collapse
|
9
|
Permana AD, Mahfud MAS, Munir M, Aries A, Rezka Putra A, Fikri A, Setiawan H, Mahendra I, Rizaludin A, Ramadhani Aziz AY, Djabir YY, Arsyad A, Harahap Y, Saputri WD, Fajarwati R, Darmawan N. A Combinatorial Approach with Microneedle Pretreatment and Thermosensitive Gel Loaded with Rivastigmine Lipid Nanoparticle Formulation Enables Brain Delivery via the Trigeminal Nerve. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68388-68406. [PMID: 39591987 DOI: 10.1021/acsami.4c16024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Alzheimer's disease (AD) often leads to dementia, causing cognitive decline and increased care needs. Rivastigmine (RV) is a key AD treatment, but its brain delivery is limited by the blood-brain barrier (BBB). Aside from oral, olfactory, and intradermal injection (i.d.) routes, the application of polymeric microneedles via the trigeminal nerve on the facial skin as a pretreatment, followed by a solid lipid nanoparticle RV-loaded thermosensitive gel (PMN-SLN-RV-TG), is an alternative to deal with the problems. This study aims to determine the optimal formula for PMN-SLN-RV-TG application and assess its brain delivery ability compared to conventional routes. The optimum SLN-RV formula had a particle size <200 nm and sustained release for 72 h, which was selected for the SLN-RV-TG formulation. SLN-RV-TG was transformed into a gel at normal skin temperature (32-37 °C), with good physical properties and nontoxic behavior. The ideal PMN formula was able to penetrate the dermal layer as an alternative to i.d. administration. Ex vivo dermatokinetics showed significant improvement of PMN-SLN-RV-TG application (p < 0.05) compared to without PMN application. In vivo pharmacokinetic studies on rats also revealed that the PMN-SLN-RV-TG had superior pharmacokinetic parameters (Cmax, AUC, t1/2, and MRT) compared to other groups (p < 0.05). Radiolabeling SLN-RV with 99mTc showed good physical properties, with a radiochemical yield of >95%. In vivo distribution studies of PMN-SLN-RV-TG application exhibited a higher brain:blood ratio than i.v. administration after 5 h, as well as being safe for the brain due to a good histological profile. These results show that PMN-SLN-RV-TG application via the trigeminal nerve on the facial skin has strong potential delivery to the brain for AD treatment.
Collapse
Affiliation(s)
- Andi Dian Permana
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi Indonesia
| | | | - Miftakul Munir
- Research Center for Radioisotope Radiopharmaceutical and Biodosimetry Technology, National Research and Innovation Agency of Indonesia, KST. BJ Habibie, South Tangerang 15314, Indonesia
| | - Arni Aries
- Research Center for Radioisotope Radiopharmaceutical and Biodosimetry Technology, National Research and Innovation Agency of Indonesia, KST. BJ Habibie, South Tangerang 15314, Indonesia
| | - Amal Rezka Putra
- Research Center for Radioisotope Radiopharmaceutical and Biodosimetry Technology, National Research and Innovation Agency of Indonesia, KST. BJ Habibie, South Tangerang 15314, Indonesia
| | - Ahsanal Fikri
- Research Center for Radioisotope Radiopharmaceutical and Biodosimetry Technology, National Research and Innovation Agency of Indonesia, KST. BJ Habibie, South Tangerang 15314, Indonesia
| | - Herlan Setiawan
- Research Center for Radioisotope Radiopharmaceutical and Biodosimetry Technology, National Research and Innovation Agency of Indonesia, KST. BJ Habibie, South Tangerang 15314, Indonesia
| | - Isa Mahendra
- Research Center for Radioisotope Radiopharmaceutical and Biodosimetry Technology, National Research and Innovation Agency of Indonesia, KST. BJ Habibie, South Tangerang 15314, Indonesia
| | - Asep Rizaludin
- Research Center for Radioisotope Radiopharmaceutical and Biodosimetry Technology, National Research and Innovation Agency of Indonesia, KST. BJ Habibie, South Tangerang 15314, Indonesia
| | | | - Yulia Yusrini Djabir
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, South Sulawesi Indonesia
| | - Aryadi Arsyad
- Faculty of Medicine, Hasanuddin University, Makassar 90245, South Sulawesi Indonesia
| | - Yahdiana Harahap
- Faculty of Pharmacy, Universitas Indonesia, Depok 16424, West Java Indonesia
| | - Wahyu Dita Saputri
- Research Center for Quantum Physics, National Research and Innovation Agency (BRIN), South Tangerang 15314, Indonesia
| | - Ria Fajarwati
- Research Center for Genetic Engineering, National Research and Innovation Agency (BRIN), Cibinong Bogor 16911, West Java Indonesia
| | - Noviyan Darmawan
- Department of Chemistry, IPB University, Bogor 16680, West Java Indonesia
| |
Collapse
|
10
|
Puranik N, Song M. Glutamate: Molecular Mechanisms and Signaling Pathway in Alzheimer's Disease, a Potential Therapeutic Target. Molecules 2024; 29:5744. [PMID: 39683904 DOI: 10.3390/molecules29235744] [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: 11/08/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Gamma-glutamate is an important excitatory neurotransmitter in the central nervous system (CNS), which plays an important role in transmitting synapses, plasticity, and other brain activities. Nevertheless, alterations in the glutamatergic signaling pathway are now accepted as a central element in Alzheimer's disease (AD) pathophysiology. One of the most prevalent types of dementia in older adults is AD, a progressive neurodegenerative illness brought on by a persistent decline in cognitive function. Since AD has been shown to be multifactorial, a variety of pharmaceutical targets may be used to treat the condition. N-methyl-D-aspartic acid receptor (NMDAR) antagonists and acetylcholinesterase inhibitors (AChEIs) are two drug classes that the Food and Drug Administration has authorized for the treatment of AD. The AChEIs approved to treat AD are galantamine, donepezil, and rivastigmine. However, memantine is the only non-competitive NMDAR antagonist that has been authorized for the treatment of AD. This review aims to outline the involvement of glutamate (GLU) at the molecular level and the signaling pathways that are associated with AD to demonstrate the drug target therapeutic potential of glutamate and its receptor. We will also consider the opinion of the leading authorities working in this area, the drawback of the existing therapeutic strategies, and the direction for the further investigation.
Collapse
Affiliation(s)
- Nidhi Puranik
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
11
|
Nevins S, McLoughlin CD, Oliveros A, Stein JB, Rashid MA, Hou Y, Jang MH, Lee KB. Nanotechnology Approaches for Prevention and Treatment of Chemotherapy-Induced Neurotoxicity, Neuropathy, and Cardiomyopathy in Breast and Ovarian Cancer Survivors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300744. [PMID: 37058079 PMCID: PMC10576016 DOI: 10.1002/smll.202300744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Indexed: 06/19/2023]
Abstract
Nanotechnology has emerged as a promising approach for the targeted delivery of therapeutic agents while improving their efficacy and safety. As a result, nanomaterial development for the selective targeting of cancers, with the possibility of treating off-target, detrimental sequelae caused by chemotherapy, is an important area of research. Breast and ovarian cancer are among the most common cancer types in women, and chemotherapy is an essential treatment modality for these diseases. However, chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy are common side effects that can affect breast and ovarian cancer survivors quality of life. Therefore, there is an urgent need to develop effective prevention and treatment strategies for these adverse effects. Nanoparticles (NPs) have extreme potential for enhancing therapeutic efficacy but require continued research to elucidate beneficial interventions for women cancer survivors. In short, nanotechnology-based approaches have emerged as promising strategies for preventing and treating chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy. NP-based drug delivery systems and therapeutics have shown potential for reducing the side effects of chemotherapeutics while improving drug efficacy. In this article, the latest nanotechnology approaches and their potential for the prevention and treatment of chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy in breast and ovarian cancer survivors are discussed.
Collapse
Affiliation(s)
- Sarah Nevins
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Callan D. McLoughlin
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Alfredo Oliveros
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Joshua B. Stein
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mohammad Abdur Rashid
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| |
Collapse
|
12
|
Shekho D, Mishra R, Kamal R, Bhatia R, Awasthi A. Breaking Barriers in Alzheimer's Disease: the Role of Advanced Drug Delivery Systems. AAPS PharmSciTech 2024; 25:207. [PMID: 39237748 DOI: 10.1208/s12249-024-02923-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024] Open
Abstract
Alzheimer's disease (AD), characterized by cognitive impairment, brain plaques, and tangles, is a global health concern affecting millions. It involves the build-up of amyloid-β (Aβ) and tau proteins, the formation of neuritic plaques and neurofibrillary tangles, cholinergic system dysfunction, genetic variations, and mitochondrial dysfunction. Various signaling pathways and metabolic processes are implicated in AD, along with numerous biomarkers used for diagnosis, risk assessment, and research. Despite these, there is no cure or effective treatment for AD. It is critically important to address this immediately to develop novel drug delivery systems (NDDS) capable of targeting the brain and delivering therapeutic agents to modulate the pathological processes of AD. This review summarizes AD, its pathogenesis, related signaling pathways, biomarkers, conventional treatments, the need for NDDS, and their application in AD treatment. It also covers preclinical, clinical, and ongoing trials, patents, and marketed AD formulations.
Collapse
Affiliation(s)
- Devank Shekho
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Ritika Mishra
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Rohit Bhatia
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India.
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
13
|
Nica I, Volovat C, Boboc D, Popa O, Ochiuz L, Vasincu D, Ghizdovat V, Agop M, Volovat CC, Lupascu Ursulescu C, Lungulescu CV, Volovat SR. A Holographic-Type Model in the Description of Polymer-Drug Delivery Processes. Pharmaceuticals (Basel) 2024; 17:541. [PMID: 38675501 PMCID: PMC11053585 DOI: 10.3390/ph17040541] [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: 03/07/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
A unitary model of drug release dynamics is proposed, assuming that the polymer-drug system can be assimilated into a multifractal mathematical object. Then, we made a description of drug release dynamics that implies, via Scale Relativity Theory, the functionality of continuous and undifferentiable curves (fractal or multifractal curves), possibly leading to holographic-like behaviors. At such a conjuncture, the Schrödinger and Madelung multifractal scenarios become compatible: in the Schrödinger multifractal scenario, various modes of drug release can be "mimicked" (via period doubling, damped oscillations, modulated and "chaotic" regimes), while the Madelung multifractal scenario involves multifractal diffusion laws (Fickian and non-Fickian diffusions). In conclusion, we propose a unitary model for describing release dynamics in polymer-drug systems. In the model proposed, the polymer-drug dynamics can be described by employing the Scale Relativity Theory in the monofractal case or also in the multifractal one.
Collapse
Affiliation(s)
- Irina Nica
- Department of Odontology-Periodontology, Fixed Prosthesis, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Constantin Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str, 700115 Iasi, Romania;
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str, 700115 Iasi, Romania;
| | - Ovidiu Popa
- Department of Emergency Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Lacramioara Ochiuz
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Decebal Vasincu
- Department of Biophysics, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Maricel Agop
- Department of Physics, “Gheorghe Asachi” Technical University of Iasi, 700050 Iasi, Romania;
- Romanian Scientists Academy, 050094 Bucharest, Romania
| | - Cristian Constantin Volovat
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.C.V.); (C.L.U.)
| | - Corina Lupascu Ursulescu
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.C.V.); (C.L.U.)
| | | | - Simona Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str, 700115 Iasi, Romania;
| |
Collapse
|
14
|
Ghosh S, Ghosh S, Sharma H, Bhaskar R, Han SS, Sinha JK. Harnessing the power of biological macromolecules in hydrogels for controlled drug release in the central nervous system: A review. Int J Biol Macromol 2024; 254:127708. [PMID: 37923043 DOI: 10.1016/j.ijbiomac.2023.127708] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
Hydrogels have immense potential in revolutionizing central nervous system (CNS) drug delivery, improving outcomes for neurological disorders. They serve as promising tools for controlled drug delivery to the CNS. Available hydrogel types include natural macromolecules (e.g., chitosan, hyaluronic acid, alginate), as well as hybrid hydrogels combining natural and synthetic polymers. Each type offers distinct advantages in terms of biocompatibility, mechanical properties, and drug release kinetics. Design and engineering considerations encompass hydrogel composition, crosslinking density, porosity, and strategies for targeted drug delivery. The review emphasizes factors affecting drug release profiles, such as hydrogel properties and formulation parameters. CNS drug delivery applications of hydrogels span a wide range of therapeutics, including small molecules, proteins and peptides, and nucleic acids. However, challenges like limited biodegradability, clearance, and effective CNS delivery persist. Incorporating 3D bioprinting technology with hydrogel-based CNS drug delivery holds the promise of highly personalized and precisely controlled therapeutic interventions for neurological disorders. The review explores emerging technologies like 3D bioprinting and nanotechnology as opportunities for enhanced precision and effectiveness in hydrogel-based CNS drug delivery. Continued research, collaboration, and technological advancements are vital for translating hydrogel-based therapies into clinical practice, benefiting patients with CNS disorders. This comprehensive review article delves into hydrogels for CNS drug delivery, addressing their types, design principles, applications, challenges, and opportunities for clinical translation.
Collapse
Affiliation(s)
- Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India; ICMR - National Institute of Nutrition, Tarnaka, Hyderabad, Telangana 500007, India
| | - Soumya Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Hitaishi Sharma
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea.
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea.
| | | |
Collapse
|
15
|
Ramalho MJ, Torres ID, Loureiro JA, Lima J, Pereira MC. Transferrin-Conjugated PLGA Nanoparticles for Co-Delivery of Temozolomide and Bortezomib to Glioblastoma Cells. ACS APPLIED NANO MATERIALS 2023; 6:14191-14203. [PMID: 37588263 PMCID: PMC10426337 DOI: 10.1021/acsanm.3c02122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Glioblastoma (GBM) represents almost half of primary brain tumors, and its standard treatment with the alkylating agent temozolomide (TMZ) is not curative. Treatment failure is partially related to intrinsic resistance mechanisms mediated by the O6-methylguanine-DNA methyltransferase (MGMT) protein, frequently overexpressed in GBM patients. Clinical trials have shown that the anticancer agent bortezomib (BTZ) can increase TMZ's therapeutic efficacy in GBM patients by downregulating MGMT expression. However, the clinical application of this therapeutic strategy has been stalled due to the high toxicity of the combined therapy. The co-delivery of TMZ and BTZ through nanoparticles (NPs) of poly(lactic-co-glycolic acid) (PLGA) is proposed in this work, aiming to explore their synergistic effect while decreasing the drug's toxicity. The developed NPs were optimized by central composite design (CCD), then further conjugated with transferrin (Tf) to enhance their GBM targeting ability by targeting the blood-brain barrier (BBB) and the cancer cells. The obtained NPs exhibited suitable GBM cell delivery features (sizes lower than 200 nm, low polydispersity, and negative surface charge) and a controlled and sustained release for 20 days. The uptake and antiproliferative effect of the developed NPs were evaluated in in vitro human GBM models. The obtained results disclosed that the NPs are rapidly taken up by the GBM cells, promoting synergistic drug effects in inhibiting tumor cell survival and proliferation. This cytotoxicity was associated with significant cellular morphological changes. Additionally, the biocompatibility of unloaded NPs was evaluated in healthy brain cells, demonstrating the safety of the nanocarrier. These findings prove that co-delivery of BTZ and TMZ in Tf-conjugated PLGA NPs is a promising approach to treat GBM, overcoming the limitations of current therapeutic strategies, such as drug resistance and increased side effects.
Collapse
Affiliation(s)
- Maria João Ramalho
- LEPABE—Laboratory
for Process Engineering, Environment, Biotechnology and Energy, Faculty
of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate
Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Inês David Torres
- LEPABE—Laboratory
for Process Engineering, Environment, Biotechnology and Energy, Faculty
of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate
Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana Angélica Loureiro
- LEPABE—Laboratory
for Process Engineering, Environment, Biotechnology and Energy, Faculty
of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate
Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Jorge Lima
- i3S—Instituto
de Investigação e Inovação em Saúde, Universidade do Porto, R. Alfredo Allen, 4200-10 135 Porto, Portugal
- Ipatimup—Instituto
de Patologia e Imunologia Molecular da Universidade do Porto, Rua Júlio Amaral de Carvalho
45, 4200-135, Porto, Portugal
- Faculty
of Medicine of Porto University, Alameda
Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Maria Carmo Pereira
- LEPABE—Laboratory
for Process Engineering, Environment, Biotechnology and Energy, Faculty
of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate
Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
16
|
Han X, Ashraf M, Tipparaju SM, Xuan W. Muscle-Brain crosstalk in cognitive impairment. Front Aging Neurosci 2023; 15:1221653. [PMID: 37577356 PMCID: PMC10413125 DOI: 10.3389/fnagi.2023.1221653] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
Sarcopenia is an age-related, involuntary loss of skeletal muscle mass and strength. Alzheimer's disease (AD) is the most common cause of dementia in elderly adults. To date, no effective cures for sarcopenia and AD are available. Physical and cognitive impairments are two major causes of disability in the elderly population, which severely decrease their quality of life and increase their economic burden. Clinically, sarcopenia is strongly associated with AD. However, the underlying factors for this association remain unknown. Mechanistic studies on muscle-brain crosstalk during cognitive impairment might shed light on new insights and novel therapeutic approaches for combating cognitive decline and AD. In this review, we summarize the latest studies emphasizing the association between sarcopenia and cognitive impairment. The underlying mechanisms involved in muscle-brain crosstalk and the potential implications of such crosstalk are discussed. Finally, future directions for drug development to improve age-related cognitive impairment and AD-related cognitive dysfunction are also explored.
Collapse
Affiliation(s)
| | | | | | - Wanling Xuan
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| |
Collapse
|
17
|
Ailioaie LM, Ailioaie C, Litscher G. Photobiomodulation in Alzheimer's Disease-A Complementary Method to State-of-the-Art Pharmaceutical Formulations and Nanomedicine? Pharmaceutics 2023; 15:916. [PMID: 36986776 PMCID: PMC10054386 DOI: 10.3390/pharmaceutics15030916] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Alzheimer's disease (AD), as a neurodegenerative disorder, usually develops slowly but gradually worsens. It accounts for approximately 70% of dementia cases worldwide, and is recognized by WHO as a public health priority. Being a multifactorial disease, the origins of AD are not satisfactorily understood. Despite huge medical expenditures and attempts to discover new pharmaceuticals or nanomedicines in recent years, there is no cure for AD and not many successful treatments are available. The current review supports introspection on the latest scientific results from the specialized literature regarding the molecular and cellular mechanisms of brain photobiomodulation, as a complementary method with implications in AD. State-of-the-art pharmaceutical formulations, development of new nanoscale materials, bionanoformulations in current applications and perspectives in AD are highlighted. Another goal of this review was to discover and to speed transition to completely new paradigms for the multi-target management of AD, to facilitate brain remodeling through new therapeutic models and high-tech medical applications with light or lasers in the integrative nanomedicine of the future. In conclusion, new insights from this interdisciplinary approach, including the latest results from photobiomodulation (PBM) applied in human clinical trials, combined with the latest nanoscale drug delivery systems to easily overcome protective brain barriers, could open new avenues to rejuvenate our central nervous system, the most fascinating and complex organ. Picosecond transcranial laser stimulation could be successfully used to cross the blood-brain barrier together with the latest nanotechnologies, nanomedicines and drug delivery systems in AD therapy. Original, smart and targeted multifunctional solutions and new nanodrugs may soon be developed to treat AD.
Collapse
Affiliation(s)
- Laura Marinela Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania
| | - Constantin Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania
| | - Gerhard Litscher
- President of ISLA (International Society for Medical Laser Applications), Research Unit of Biomedical Engineering in Anesthesia and Intensive Care Medicine, Research Unit for Complementary and Integrative Laser Medicine, Traditional Chinese Medicine (TCM) Research Center Graz, Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 39, 8036 Graz, Austria
| |
Collapse
|
18
|
Caffeic Acid-Loaded Liposomes Functionalized with Transferrin for Alzheimer's Disease Therapy. Colloids Surf B Biointerfaces 2023; 225:113270. [PMID: 36996633 DOI: 10.1016/j.colsurfb.2023.113270] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/02/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023]
Abstract
Alzheimer's disease (AD) is an incurable neurological illness and the leading cause of dementia, characterized by amyloid β (Aβ) fibril deposits. Caffeic acid (CA) has demonstrated potential value for AD therapy due to its anti-amyloidogenic, anti-inflammatory, and antioxidant properties. However, its chemical instability and limited bioavailability limit its therapeutic potential in vivo. Herein, liposomes loading CA were produced by distinct techniques. Taking advantage of the overexpression of transferrin (Tf) receptors in brain endothelial cells, Tf was conjugated to the liposomes' surface to direct the CA-loaded nanoparticles (NPs) to the blood-brain barrier (BBB). The optimized Tf-modified NPs exhibited a mean size of around 140 nm, a polydispersity index lower than 0.2, and a neutral surface charge, being appropriate for drug delivery. The Tf-functionalized liposomes showed suitable encapsulation efficiency and physical stability for at least 2 months. Furthermore, in simulated physiological settings, the NPs ensured the sustained release of CA for 8 days. The anti-amyloidogenic efficacy of the optimized drug delivery system (DDS) was investigated. The data show that CA-loaded Tf-functionalized liposomes are capable of preventing Aβ aggregation and fibril formation, and disaggregating mature fibrils. Hence, the proposed brain-targeted DDS may be a potential strategy for preventing and treating AD. Future studies in animal models of AD will be valuable to validate the therapeutic efficacy of the optimized nanosystem.
Collapse
|
19
|
Intrathecal Pseudodelivery of Drugs in the Therapy of Neurodegenerative Diseases: Rationale, Basis and Potential Applications. Pharmaceutics 2023; 15:pharmaceutics15030768. [PMID: 36986629 PMCID: PMC10059785 DOI: 10.3390/pharmaceutics15030768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Intrathecal pseudodelivery of drugs is a novel route to administer medications to treat neurodegenerative diseases based on the CSF-sink therapeutic strategy by means of implantable devices. While the development of this therapy is still in the preclinical stage, it offers promising advantages over traditional routes of drug delivery. In this paper, we describe the rationale of this system and provide a technical report on the mechanism of action, that relies on the use of nanoporous membranes enabling selective molecular permeability. On one side, the membranes do not permit the crossing of certain drugs; whereas, on the other side, they permit the crossing of target molecules present in the CSF. Target molecules, by binding drugs inside the system, are retained or cleaved and subsequently eliminated from the central nervous system. Finally, we provide a list of potential indications, the respective molecular targets, and the proposed therapeutic agents.
Collapse
|
20
|
Wang H, Huber CC, Li XP. Mesenchymal and Neural Stem Cell-Derived Exosomes in Treating Alzheimer's Disease. Bioengineering (Basel) 2023; 10:253. [PMID: 36829747 PMCID: PMC9952071 DOI: 10.3390/bioengineering10020253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/17/2023] Open
Abstract
As the most common form of dementia and a progressive neurodegenerative disorder, Alzheimer's disease (AD) affects over 10% world population with age 65 and older. The disease is neuropathologically associated with progressive loss of neurons and synapses in specific brain regions, deposition of amyloid plaques and neurofibrillary tangles, neuroinflammation, blood-brain barrier (BBB) breakdown, mitochondrial dysfunction, and oxidative stress. Despite the intensive effort, there is still no cure for the disorder. Stem cell-derived exosomes hold great promise in treating various diseases, including AD, as they contain a variety of anti-apoptotic, anti-inflammatory, and antioxidant components. Moreover, stem cell-derived exosomes also promote neurogenesis and angiogenesis and can repair damaged BBB. In this review, we will first outline the major neuropathological features associated with AD; subsequently, a discussion of stem cells, stem cell-secreted exosomes, and the major exosome isolation methods will follow. We will then summarize the recent data involving the use of mesenchymal stem cell- or neural stem cell-derived exosomes in treating AD. Finally, we will briefly discuss the challenges, perspectives, and clinical trials using stem cell-derived exosomes for AD therapy.
Collapse
Affiliation(s)
- Hongmin Wang
- Division of Basic Biomedical Sciences and Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | | | | |
Collapse
|
21
|
Mostovaya O, Shiabiev I, Pysin D, Stanavaya A, Abashkin V, Shcharbin D, Padnya P, Stoikov I. PAMAM-Calix-Dendrimers: Second Generation Synthesis, Fluorescent Properties and Catecholamines Binding. Pharmaceutics 2022; 14:pharmaceutics14122748. [PMID: 36559243 PMCID: PMC9781744 DOI: 10.3390/pharmaceutics14122748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
A convenient method for the synthesis of the second generation of PAMAM dendrimers based on a p-tert-butylthiacalix[4]arene core in cone, partial cone and 1,3-alternate conformations was developed. Unusual fluorescence of the obtained PAMAM-calix-dendrimers has been found and explained. The binding ability of the synthesized dendrimers toward catecholamines (dopamine, L-adrenaline and L-noradrenaline) was shown by UV-Vis, fluorescence, 1D and 2D NMR spectroscopy and the binding constants (logKa 3.85-4.74) calculated. As was shown, the PAMAM-calix-dendrimers bind catecholamines by the internal cavities. All the studied hormones were most efficiently bound by the dendrimers bearing a macrocyclic core in 1,3-alternate conformation. The size of the formed supramolecular systems of dendrimer/catecholamine was established by the DLS method. A decrease in hemolytic activity of the PAMAM-calix-dendrimers with an increase in the generation number of a dendrimer was shown for the dendrimers with a core in 1,3-alternate conformation. The prospects for the use of the synthesized dendrimers with the macrocyclic core as drug delivery agents were discussed.
Collapse
Affiliation(s)
- Olga Mostovaya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russia
- Correspondence: (O.M.); (I.S.); Tel.: +7-843-233-7241 (I.S.)
| | - Igor Shiabiev
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russia
| | - Dmitry Pysin
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russia
| | - Alesia Stanavaya
- Institute of Biophysics and Cell Engineering of NASB, 27 Akademicheskaya Street, 220072 Minsk, Belarus
| | - Viktar Abashkin
- Institute of Biophysics and Cell Engineering of NASB, 27 Akademicheskaya Street, 220072 Minsk, Belarus
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, 27 Akademicheskaya Street, 220072 Minsk, Belarus
| | - Pavel Padnya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russia
| | - Ivan Stoikov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russia
- Federal Center for Toxicological, Radiation and Biological Safety, 2 Nauchny Gorodok Street, Kazan 420075, Russia
- Correspondence: (O.M.); (I.S.); Tel.: +7-843-233-7241 (I.S.)
| |
Collapse
|