1
|
Zohora FT, Pathmanathan R, Chowdhury EH. Application of Strontium Chloride Hexahydrate to Synthesize Strontium-Substituted Carbonate Apatite as a pH-Sensitive, Biologically Safe, and Highly Efficient siRNA Nanocarrier. ACS APPLIED BIO MATERIALS 2024. [PMID: 39723844 DOI: 10.1021/acsabm.4c01319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Naked siRNAs are sensitive to enzymatic degradation, phagocytic entrapment, quick renal excretion, membrane impermeability, endosomal escape, and off-target effects. Designing a safe and efficient nanocarrier for siRNA delivery to the target site without toxicity remains a significant hurdle in gene therapy. CA is a unique derivative of hydroxyapatite and a highly pH-sensitive nanocarrier with strong particle aggregation and a high polydispersity index. Strontium (Sr2+), a group two divalent metal in the periodic table, has been reported for substituting calcium (Ca2+) ions from the apatite lattice and limiting particle growth/aggregation. This study used strontium chloride hexahydrate (SrCl2·6H2O) salt to develop a Sr-substituted CA (Sr-CA) nanocarrier with ∼30 nm size, spherical shape, less aggregation, homodispersity, and a fair anionic charge. Sr-CA demonstrated a large surface area-to-volume ratio, an improved cargo loading efficiency, and enhanced cellular uptake in HEK-293 cells. Moreover, Sr-CA is a pH-responsive nanocarrier responsible for its long physiological stability, efficient endosomal escape, and optimal cargo delivery within cells. These NPs have differential effects on MAPK1, MAP2K4, PIK3Ca, CAMK4, and p53 gene expression in HEK-293 cells without showing any significant cytotoxicity in cell growth properties. Gene silencing by Sr-CA-mediated siRNA delivery against MAPK1, MAP2K4, PIK3Ca, and CAMK4 genes significantly decreased the level of target gene expression and cell survival, demonstrating successful intracellular siRNA delivery in HEK-293 cells. Additionally, biocompatibility testing confirmed the biological safety of the Sr-CA nanocarrier in mice. These findings suggest that Sr-CA nanocarriers are a promising siRNA delivery system, combining high efficiency with pH-sensitive release and excellent biocompatibility, making them a viable option for future therapeutic applications.
Collapse
Affiliation(s)
- Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Rajadurai Pathmanathan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
- Daffodil International University, Daffodil Smart City, Birulia 1216, Bangladesh
- Nanoflex LLC, 31756 Broadwater Avenue, Leesburg, Florida 34748, United States
| |
Collapse
|
2
|
Morsy HM, Zaky MY, Yassin NYS, Khalifa AYZ. Nanoparticle-based flavonoid therapeutics: Pioneering biomedical applications in antioxidants, cancer treatment, cardiovascular health, neuroprotection, and cosmeceuticals. Int J Pharm 2024; 670:125135. [PMID: 39732216 DOI: 10.1016/j.ijpharm.2024.125135] [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: 09/05/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Flavonoids, a type of natural polyphenolic molecule, have garnered significant research interest due to their ubiquitous nature and diverse biological activities, including antioxidant, anti-inflammatory, and anticancer effects, making them appealing to various scientific disciplines. In this regard, the use of a flavonoid nanoparticle delivery system is to overcome low bioavailability, bioactivity, poor aqueous solubility, systemic absorption, and intensive metabolism. Therefore, this review summarizes the classification of nanoparticles (liposomes, polymeric, and solid lipid nanoparticles) and the advantages of using nanoparticle-flavonoid formulations to boost flavonoid bioavailability. Moreover, this review illustrated the pioneering biomedical applications of nanoparticle-based flavonoid therapeutics, as well as safety and toxicity considerations of using a flavonoid nanoparticle delivery system.
Collapse
Affiliation(s)
- Hadeer M Morsy
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Mohamed Y Zaky
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt.
| | - Nour Y S Yassin
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Ashraf Y Z Khalifa
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia.
| |
Collapse
|
3
|
Razack SA, Kim YE, Kang HW. κ-carrageenan - Gelatin hydrogel embedding carvacrol loaded gold nanobipyramids for treating prostate cancer via fractionated photothermal-chemotherapy. Int J Biol Macromol 2024:138974. [PMID: 39710028 DOI: 10.1016/j.ijbiomac.2024.138974] [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: 09/30/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Conventional treatment of prostate cancer need more specificity, and higher efficiency. The present work is the first attempt to utilize hydrogel-loaded carvacrol-based chemotherapy with fractionated photothermal therapy (F-PTT) using a 635 nm laser for its treatment. Gold nanobipyramids (AuNBPs) were used as drug carrier and photosensitizer. A marine-derived hydrogel (AuNBP-HG) was fabricated and physicochemically characterized. A 635 nm NIR light was used for the fractionated PTT in three cycles of treatment on different days at varying power densities for 5 min. The efficiency of single and dual treatments was experimented in vitro and in vivo. The results showed that the dual therapy imparted a better effect than monotherapy. The AuNBP-CVL synthesized was nearly 50 nm and efficiently loaded within AuNBP-HG, displaying thermal responsiveness, a good sol-gel transition, a controlled drug release rate, and high stability. In vitro evaluation demonstrated that the F-PTT evinced stability during temperature rise without damaging healthy surrounding tissue. In vivo study revealed that a stable temperature rise up to 60 °C by the 635 nm NIR light due to photosensitization of AuNBP and sustained CVL release from AuNBP-HG into the tumor microenvironment exhibited successful thermal ablation of the cancer cells. Histological analysis confirmed that the uniform suspension of AuNBP within cancer tissues might have enhanced tumor cell lysis during the dual therapy. Western blot analysis exhibited that the cellular death could result from the upregulation of pJNK and p53 protein. In conclusion, the current study illustrated that dual therapy could be a feasible alternative to non-target specific and invasive traditional therapies against prostate cancer.
Collapse
Affiliation(s)
- Sirajunnisa Abdul Razack
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
| | - Yeong Eun Kim
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea
| | - Hyun Wook Kang
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea; Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information, Pukyong National University, Busan, Republic of Korea.
| |
Collapse
|
4
|
Sutcliffe R, Doherty CPA, Morgan HP, Dunne NJ, McCarthy HO. Strategies for the design of biomimetic cell-penetrating peptides using AI-driven in silico tools for drug delivery. BIOMATERIALS ADVANCES 2024; 169:214153. [PMID: 39705787 DOI: 10.1016/j.bioadv.2024.214153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/08/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Cell-penetrating peptides (CPP) have gained rapid attention over the last 25 years; this is attributed to their versatility, customisation, and 'Trojan horse' delivery that evades the immune system. However, the current CPP rational design process is limited, as it requires several rounds of peptide synthesis, prediction and wet-lab validation, which is expensive, time-consuming and requires extensive knowledge in peptide chemistry. Artificial intelligence (AI) has emerged as a promising alternative which can augment the design process, for example by determining physiochemical characteristics, secondary structure, solvent accessibility, disorder and flexibility, as well as predicting in vivo behaviour such as toxicity and peptidase degradation. Other more recent tools utilise supervised machine learning (ML) to predict the penetrative ability of an amino acid sequence. The use of AI in the CPP design process has the potential to reduce development costs and increase the chances of success with respect to delivery. This review provides a survey of in silico tools and AI platforms which can be utilised in the design process, and the key features that should be taken into consideration when designing next generation CPPs.
Collapse
Affiliation(s)
- Rebecca Sutcliffe
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland
| | - Ciaran P A Doherty
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; Antigenesis Biologics, Crossgar, Northern Ireland, United Kingdom of Great Britain and Northern Ireland
| | - Hugh P Morgan
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; Antigenesis Biologics, Crossgar, Northern Ireland, United Kingdom of Great Britain and Northern Ireland
| | - Nicholas J Dunne
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland; School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
5
|
Klika K, Han J, Busse MS, Soloshonok VA, Javahershenas R, Vanhaecke F, Makarem A. Inductively Coupled Plasma-Mass Spectrometry (ICP-MS): An Emerging Tool in Radiopharmaceutical Science. J Am Chem Soc 2024; 146:30717-30727. [PMID: 39478417 PMCID: PMC11565647 DOI: 10.1021/jacs.4c12254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/14/2024]
Abstract
Although radioactive experiments are necessary in radiopharmaceutical drug discovery and theranostic cancer research, they are expensive, require special facilities, and face certain restrictions. Thus, finding techniques not involving radioactivity is highly beneficial for minimizing these disadvantages in such research. In this regard, methods using inductively coupled plasma-mass spectrometry (ICP-MS) have emerged as viable alternatives to traditional radioactive approaches. Despite its potential, practical applications of ICP-MS in radiopharmaceutical cancer research have only emerged in recent years. This Perspective focuses on the development and implementation of nonradioactive ICP-MS-based assays in radiopharmaceutical research and aims to inspire future research efforts in this area.
Collapse
Affiliation(s)
- Karel
D. Klika
- Molecular
Structure Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jianlin Han
- College
of Chemical Engineering, Nanjing Forestry
University, 210037 Nanjing, China
| | - Marvin S. Busse
- Institute
of Pharmacy, University of Hamburg, 20146 Hamburg, Germany
| | - Vadim A. Soloshonok
- Department
of Organic Chemistry I, University of the
Basque Country, 20018 San Sebastián, Spain
- IKERBASQUE,
Basque Foundation for Science, 48009 Bilbao, Spain
| | - Ramin Javahershenas
- Department
of Organic Chemistry, Faculty of Chemistry, Urmia University, 57179-44514 Urmia, Iran
| | - Frank Vanhaecke
- Atomic
and Mass Spectrometry − A&MS Research Unit, Department
of Chemistry, Ghent University, 9000 Ghent, Belgium
| | - Ata Makarem
- Institute
of Pharmacy, University of Hamburg, 20146 Hamburg, Germany
| |
Collapse
|
6
|
Zhang LK, Li Y, Zhai L, Tang Y, Jiao Y, Mei Y, Yang R, You R, Yin L, Ni H, Ge J, Guan YQ. Natural Phycocyanin/Paclitaxel Micelle Delivery of Therapeutic P53 to Activate Apoptosis for HER2 or ER Positive Breast Cancer Therapy. ACS Biomater Sci Eng 2024; 10:6995-7004. [PMID: 39390952 DOI: 10.1021/acsbiomaterials.4c00756] [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: 10/12/2024]
Abstract
The P53 gene is commonly mutated in breast cancer, protein based the gene as anticancer drugs could provide efficient and stable advantages by restoring the function of the wild-type P53 protein. In this study, we describe the creation and utilization of a micelle composed by natural phycocyanin and paclitaxel and grafting anti-HER2 (PPH), which effectively packages and transports recombinant P53 protein with anti-ER (PE), resulting in a new entity designated as PE@PPH, to address localization obstacles and modify cellular tropism to the cell membrane or nucleus. The results indicate that PE@PPH has strong antitumor properties, even at low doses of PTX both in vitro and in vivo. These findings suggest that PE@PPH could be an enhancing micelle for delivering therapeutic proteins and promoting protein functional recovery, particularly in addressing the challenges posed by tumor heterogeneity in breast cancer.
Collapse
Affiliation(s)
- Ling-Kun Zhang
- School of Life Science, South China Normal University, Guangzhou 510631, China
- School of Engineering, Westlake University, Hangzhou 310030, China
- Institute of Advanced Technology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Yuan Li
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Limin Zhai
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yunzhi Tang
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yuxuan Jiao
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Yitong Mei
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Runcai Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Rong You
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Liang Yin
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - He Ni
- School of Life Science, South China Normal University, Guangzhou 510631, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, China
| | - Yan-Qing Guan
- School of Life Science, South China Normal University, Guangzhou 510631, China
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
7
|
Pashirova T, Shaihutdinova Z, Tatarinov D, Titova A, Malanyeva A, Vasileva O, Gabdurakhmanov K, Dudnikov S, Schopfer LM, Lockridge O, Masson P. Pharmacokinetics and fate of free and encapsulated IRD800CW-labelled human BChE intravenously administered in mice. Int J Biol Macromol 2024; 282:137305. [PMID: 39515732 DOI: 10.1016/j.ijbiomac.2024.137305] [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: 08/06/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Human butyrylcholinesterase (BChE) is an efficient bioscavenger of toxicants. Highly purified BChE was labelled with the near infrared fluorescent IRDye800CW. The goal was to determine the pharmacokinetics and fate of enzyme in mice. BChE-IRDye800CW was encapsulated in polyethylene glycol-polypropylene sulfide-based spherical polymersome nanoreactors with the following characteristics: 140 nm diameter, ξ = -6 mV, PDI ≤ 0.2, 1 year stability. Encapsulation did not alter the functional properties of BChE. Free and encapsulated enzyme were injected intravenously to CD-1 mice (single dose of enzyme 1.5 mg/kg and PEG-PPS polymersomes 25 mg/kg) and were analyzed for 8 days using an in vivo imaging system. Results showed that the pharmacokinetic distribution α-phase of encapsulated BChE (t1/2 = 17.6 h) was longer than for free enzyme (t1/2 = 6.6 h). The mean half-time for elimination β-phase was 2-time longer for encapsulated enzyme than for free enzyme (150 vs 72 h). Transient changes in infrared fluorescence in organs showed that BChE is eliminated from liver. However, free and encapsulated enzymes were cleared via different pathways. This first study of pharmacokinetics and fate of BChE encapsulated in polymersomes initiates research of new formulations of bioscavengers aimed at increasing the residence time of enzymes in the blood stream.
Collapse
Affiliation(s)
- Tatiana Pashirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation; Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation.
| | - Zukhra Shaihutdinova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation; Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation
| | - Dmitry Tatarinov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Str. 8, 420088 Kazan, Russian Federation
| | - Angelina Titova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Albina Malanyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Olga Vasileva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Kamil Gabdurakhmanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | - Sergei Dudnikov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation
| | | | - Oksana Lockridge
- University of Nebraska Medical Center, Eppley Institute, Omaha, NE, USA
| | - Patrick Masson
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, 18 Kremlyovskaya St., Russian Federation.
| |
Collapse
|
8
|
Macone A, Cappelletti C, Incocciati A, Piacentini R, Botta S, Boffi A, Bonamore A. Challenges in Exploiting Human H Ferritin Nanoparticles for Drug Delivery: Navigating Physiological Constraints. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2016. [PMID: 39541599 DOI: 10.1002/wnan.2016] [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: 06/24/2024] [Revised: 10/14/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Over the past two decades, ferritin has emerged as a promising nanoparticle for drug delivery, catalyzing the development of numerous prototypes capable of encapsulating a wide array of therapeutic agents. These ferritin-based nanoparticles exhibit selectivity for various molecular targets and are distinguished by their potential biocompatibility, unique symmetrical structure, and highly controlled size. The hollow interior of ferritin nanoparticles allows for efficient encapsulation of diverse therapeutic agents, enhancing their delivery and effectiveness. Despite these promising features, the anticipated clinical advancements have yet to be fully realized. As a physiological protein with a central role in both health and disease, ferritin can exert unexpected effects on physiology when employed as a drug delivery system. Many studies have not thoroughly evaluated the pharmacokinetic properties of the ferritin protein shell when administered in vivo, overlooking crucial aspects such as biodistribution, clearance, cellular trafficking, and immune response. Addressing these challenges is crucial for achieving the desired transition from bench to bedside. Biodistribution studies need to account for ferritin's natural accumulation in specific organs (liver, spleen, and kidneys), which may lead to off-target effects. Moreover, the mechanisms of clearance and cellular trafficking must be elucidated to optimize the delivery and reduce potential toxicity of ferritin nanoparticles. Additionally, understanding the immune response elicited by exogenous ferritin is essential to mitigate adverse reactions and enhance therapeutic efficacy. A comprehensive understanding of these physiological constraints, along with innovative solutions, is essential to fully realize the therapeutic potential of ferritin nanoparticles paving the way for their successful clinical translation.
Collapse
Affiliation(s)
- Alberto Macone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Chiara Cappelletti
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessio Incocciati
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Roberta Piacentini
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Sofia Botta
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alberto Boffi
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Alessandra Bonamore
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
9
|
Wang H, Wang X, Wang L, Wang H, Zhang Y. Exploiting lignin-based nanomaterials for enhanced anticancer therapy: A comprehensive review and future direction. Int J Biol Macromol 2024; 281:136266. [PMID: 39366596 DOI: 10.1016/j.ijbiomac.2024.136266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/18/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Lignin, a renewable and abundant natural polymer, has emerged as a promising candidate for anticancer therapy due to its unique properties and biocompatibility. This review provides a comprehensive overview of recent advancements in the utilization of lignin-based nanomaterials for enhancing anticancer drug delivery and therapeutic outcomes. A detailed examination of the literature reveals several synthesis methods, including nanoprecipitation, microemulsion, and solvent exchange, which produce lignin nanoparticles with improved drug solubility and bioavailability. The anticancer mechanisms of lignin nanoparticles, such as the generation of reactive oxygen species (ROS), induction of apoptosis, and enhanced cellular uptake, are also explored. Lignin nanoparticles loaded with drugs like curcumin, doxorubicin, camptothecin, and resveratrol have demonstrated the ability to improve drug efficacy, selectively target cancer cells, overcome multidrug resistance, and minimize toxicity in both in vitro and in vivo studies. These nanoparticles have shown significant potential in suppressing tumor growth, inducing cell death through apoptotic pathways, and enhancing the synergistic effects of combination therapies, such as chemo-phototherapy. Future research directions include optimizing lignin nanoparticle formulations for clinical applications, refining targeted delivery mechanisms to cancer cells, and conducting thorough biocompatibility and toxicity assessments. Overall, this review highlights the significant progress made in utilizing lignin-based nanomaterials for cancer therapy and outlines promising areas for further exploration in this rapidly evolving field.
Collapse
Affiliation(s)
- Haoyu Wang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China; Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaoyang Wang
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Long Wang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China
| | - Haifan Wang
- Department of Orthopedics, The Second Affiliated Hospital, Xi'an, Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yuxing Zhang
- Biomedical Research Center of Xijing University, Xi'an, Shaanxi 710123, China.
| |
Collapse
|
10
|
Subhasri D, Leena MM, Moses JA, Anandharamakrishnan C. Factors affecting the fate of nanoencapsulates post administration. Crit Rev Food Sci Nutr 2024; 64:11949-11973. [PMID: 37599624 DOI: 10.1080/10408398.2023.2245462] [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: 08/22/2023]
Abstract
Nanoencapsulation has found numerous applications in the food and nutraceutical industries. Micro and nanoencapsulated forms of bioactives have proven benefits in terms of stability, release, and performance in the body. However, the encapsulated ingredient is often subjected to a wide range of processing conditions and this is followed by storage, consumption, and transit along the gastrointestinal tract. A strong understanding of the fate of nanoencapsulates in the biological system is mandatory as it provides valuable insights for ingredient selection, formulation, and application. In addition to their efficacy, there is also the need to assess the safety of ingested nanoencapsulates. Given the rising research and commercial focus of this subject, this review provides a strong focus on their interaction factors and mechanisms, highlighting their prospective biological fate. This review also covers various approaches to studying the fate of nanoencapsulates in the body. Also, with emphasis on the overall scope, the need for a new advanced integrated common methodology to evaluate the fate of nanoencapsulates post-administration is discussed.
Collapse
Affiliation(s)
- D Subhasri
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology Entrepreneurship and Management - Thanjavur, Ministry of Food Processing Industries, Government of India, Thanjavur, India
| | - M Maria Leena
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology Entrepreneurship and Management - Thanjavur, Ministry of Food Processing Industries, Government of India, Thanjavur, India
- Department of Biotechnology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Tiruchirappalli, India
| | - J A Moses
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology Entrepreneurship and Management - Thanjavur, Ministry of Food Processing Industries, Government of India, Thanjavur, India
| | - C Anandharamakrishnan
- Computational Modeling and Nanoscale Processing Unit, National Institute of Food Technology Entrepreneurship and Management - Thanjavur, Ministry of Food Processing Industries, Government of India, Thanjavur, India
- CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Ministry of Science and Technology, Government of India, Industrial Estate PO, Thiruvananthapuram, INDIA
| |
Collapse
|
11
|
Chang X, Liu J, Li Y, Li W. Pluronic F127-Complexed PEGylated Poly(glutamic acid)-Cisplatin Nanomedicine for Enhanced Glioblastoma Therapy. Macromol Rapid Commun 2024; 45:e2400662. [PMID: 39264576 DOI: 10.1002/marc.202400662] [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] [Indexed: 09/13/2024]
Abstract
Glioblastoma is one of the most aggressive and treatment-resistant forms of primary brain cancer, posing significant challenges in effective therapy. This study aimed to enhance the effectiveness of glioblastoma therapy by developing a unique nanomedicine composed of Pluronic F127-complexed PEGylated poly(glutamic acid)-cisplatin (PLG-PEG/PF127-CDDP). PLG-PEG/PF127-CDDP demonstrated an optimal size of 133.97 ± 12.60 nm, facilitating efficient cell uptake by GL261 glioma cells. In vitro studies showed significant cytotoxicity against glioma cells with a half-maximal (50%) inhibitory concentration (IC50) of 12.61 µg mL-1 at 48 h and a 72.53% ± 1.89% reduction in cell invasion. Furthermore, PLG-PEG/PF127-CDDP prolonged the circulation half-life of cisplatin to 9.75 h in vivo, leading to a more than 50% reduction in tumor size on day 16 post-treatment initiation in a murine model of glioma. The treatment significantly elevated lactate levels in GL261 cells, indicating enhanced metabolic disruption. Therefore, PLG-PEG/PF127-CDDP offers a promising approach for glioblastoma therapy due to its effects on improving drug delivery efficiency, therapeutic outcomes, and safety while minimizing systemic side effects. This work underscores the potential of polymer-based nanomedicines in overcoming the challenges of treating brain tumors, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Xiaoyu Chang
- Department of Neurosurgery, the First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
| | - Jiaxue Liu
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, 5 Jilin Street, Jilin, 132000, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Yunqian Li
- Department of Neurosurgery, the First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
| | - Wenliang Li
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, 5 Jilin Street, Jilin, 132000, P. R. China
| |
Collapse
|
12
|
Yuan S, Zhu L, Luo Y, Chen X, Jing H, Wang J, Su X, Liang M, Zhuang Z. Igniting tumour microenvironment in triple-negative breast cancer using a mannose/hyaluronic acid dual-coated Ganoderma polysaccharide-superparamagnetic iron oxide nanocomplex for combinational therapies. J Drug Target 2024:1-16. [PMID: 39470031 DOI: 10.1080/1061186x.2024.2408721] [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: 07/16/2024] [Revised: 08/31/2024] [Accepted: 09/21/2024] [Indexed: 10/30/2024]
Abstract
Eliciting tumour microenvironment (TME) activation in triple-negative breast cancer (TNBC) is crucial for effective anti-tumour therapies. The aim of this study is to employ pharmaceutical approaches to precisely deliver Ganoderma polysaccharide (GPS) to tumour sites, thereby enhancing TME activation. We first established a direct link between the accumulation of GPS within tumours and its efficacy in the TME activation. Building upon this insight, we then engineered a mannose/hyaluronic acid dual-coated GPS-loaded superparamagnetic iron oxide nanocomplex (Man/HA/GPS-SPIONs) with a particle size of 33.8 ± 1.6 nm and a zeta potential of -22.4 ± 3.5 mV, capable of precise tumour accumulation through magnet-assisted targeting and internalisation by tumour-associated macrophages (TAMs) and tumour cells, facilitated by dual ligand modification. In vitro, Man/HA/GPS-SPIONs effectively induced M1 polarisation of macrophages (CD86+ cells: 38.6 ± 2.8%), curbed 4T1 cell proliferation (viability: 47.3 ± 2.9%) and heightened Th1 cytokine release. Significantly, in vivo, Man/HA/GPS-SPIONs notably suppressed tumour growth (tumour index: 0.048 ± 0.005), fostered M1 polarisation of TAMs (CD45+F4/80+CD86+ cells: 26.1 ± 7.2%), consequently bolstering intratumoural T cytotoxic cells. This enhancement was intricately tied to the efficient co-delivery of GPS and iron ions to the tumours, made possible by the Man/HA/GPS-SPIONs delivery system. The synergistic effects with paclitaxel (PTX, inhibition rate: 61.2 ± 4.3%) and PD-1 inhibitors (inhibition rate: 69.8 ± 7.6%) underscored the translational potential of this approach. By harnessing a well-conceived iron-based drug delivery strategy, this study amplifies the tumour immune modulatory potential of natural polysaccharides, offering insightful guidance for interventions in the TME and synergistic therapies.
Collapse
Affiliation(s)
- Shaofei Yuan
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, P.R. China
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Linjia Zhu
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Yi Luo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaoqiang Chen
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Haibo Jing
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Jiaqi Wang
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Xiangyu Su
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Meizhen Liang
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, P.R. China
| |
Collapse
|
13
|
Fusco L, Gazzi A, Giro L, Schefer RB, D'Almeida SM, Cagliani R, Zoccheddu M, Uyar R, Besbinar Ö, Çelik D, Yilmazer A, Mitrano DM, Orecchioni M, Delogu LG. Nanoplastics: Immune Impact, Detection, and Internalization after Human Blood Exposure by Single-Cell Mass Cytometry. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2413413. [PMID: 39449193 DOI: 10.1002/adma.202413413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/07/2024] [Indexed: 10/26/2024]
Abstract
The increasing exposure to nanoplastics (NPs) raises significant concerns for human health, primarily due to their potential bioaccumulative properties. While NPs have recently been detected in human blood, their interactions with specific immune cell subtypes and their impact on immune regulation remain unclear. In this proof-of-concept study, model palladium-doped polystyrene NPs (PS-Pd NPs) are utilized to enable single-cell mass cytometry (CyTOF) detection. The size-dependent impact of carboxylate polystyrene NPs (50-200 nm) is investigated across 15 primary immune cell subpopulations using CyTOF. By taking advantage of Pd-doping for detecting PS-Pd NPs, this work evaluates their impact on human immune-cells at the single-cell level following blood exposure. This work traces PS-Pd NPs in 37 primary immune-cell subpopulations from human blood, quantifying the palladium atom count per cell by CyTOF while simultaneously assessing the impact of PS-Pd NPs on cell viability, functionality, and uptake. These results demonstrate that NPs can interact with, interfere with, and translocate into several immune cell subpopulations after exposure. In vivo distribution experiments in mice further confirmed their accumulation in immune cells within the liver, blood, and spleen, particularly in monocytes, macrophages, and dendritic cells. These findings provide valuable insights into the impact of NPs on human health.
Collapse
Affiliation(s)
- Laura Fusco
- ImmuneNano-lab, Department of Biomedical Sciences, University of Padua, Padua, 35131, Italy
| | - Arianna Gazzi
- ImmuneNano-lab, Department of Biomedical Sciences, University of Padua, Padua, 35131, Italy
| | - Linda Giro
- ImmuneNano-lab, Department of Biomedical Sciences, University of Padua, Padua, 35131, Italy
- Immunology Center of Georgia, Augusta University, Augusta, 30912, United States
| | - Roman B Schefer
- Department of Environmental Systems Science, ETH Zürich, Zürich, 8092, Switzerland
| | | | - Roberta Cagliani
- ImmuneNano-lab, Department of Biomedical Sciences, University of Padua, Padua, 35131, Italy
| | - Martina Zoccheddu
- Immunology Center of Georgia, Augusta University, Augusta, 30912, United States
- Georgia Cancer Center Integrated Genomics core, Augusta University, Augusta, 30912, United States
| | - Recep Uyar
- Stem Cell Institute, Ankara University, Ankara, 06100, Turkey
| | - Ömur Besbinar
- Stem Cell Institute, Ankara University, Ankara, 06100, Turkey
| | - Doğantan Çelik
- Stem Cell Institute, Ankara University, Ankara, 06100, Turkey
| | - Acelya Yilmazer
- Stem Cell Institute, Ankara University, Ankara, 06100, Turkey
- Department of Biomedical Engineering, Faculty of Engineering, Ankara University, Ankara, 06100, Turkey
| | - Denise M Mitrano
- Department of Environmental Systems Science, ETH Zürich, Zürich, 8092, Switzerland
| | - Marco Orecchioni
- Immunology Center of Georgia, Augusta University, Augusta, 30912, United States
- Department of Pharmacology & Toxicology, Augusta University, Augusta, 30912, United States
| | - Lucia Gemma Delogu
- ImmuneNano-lab, Department of Biomedical Sciences, University of Padua, Padua, 35131, Italy
- Department of Biological Sciences, Khalifa University of Science & Technology, Abu Dhabi, 127788, UAE
| |
Collapse
|
14
|
Benyettou F, Khair M, Prakasam T, Varghese S, Matouk Z, Alkaabi M, Pena-Sánchez P, Boitet M, AbdulHalim R, Sharma SK, Ghemrawi R, Thomas S, Whelan J, Pasricha R, Jagannathan R, Gándara F, Trabolsi A. cRGD-Peptide Modified Covalent Organic Frameworks for Precision Chemotherapy in Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56676-56695. [PMID: 39267454 PMCID: PMC11503616 DOI: 10.1021/acsami.4c10812] [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: 07/01/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024]
Abstract
This study presents the use of nanoscale covalent organic frameworks (nCOFs) conjugated with tumor-targeting peptides for the targeted therapy of triple-negative breast cancer (TNBC). While peptides have previously been used for targeted delivery, their conjugation with COFs represents an innovative approach in this field. In particular, we have developed alkyne-functionalized nCOFs chemically modified with cyclic RGD peptides (Alkyn-nCOF-cRGD). This configuration is designed to specifically target αvβ3 integrins that are overexpressed in TNBC cells. These nCOFs exhibit excellent biocompatibility and are engineered to selectively disintegrate under acidic conditions, allowing for precise and localized drug release in tumor environment. Doxorubicin, a chemotherapeutic agent, has been encapsulated in these nCOFs with high loading efficiency. The therapeutic potential of Alkyn-nCOF-cRGD has been demonstrated in vitro and in vivo models. It shows significantly improved drug uptake and targeted cell death in TNBC, highlighting the efficacy of receptor-mediated endocytosis and pH-controlled drug release. This strategy leverages the unique properties of nCOFs with targeted drug delivery to achieve significant advances in personalized cancer therapy and set a new standard for precision chemotherapeutic delivery.
Collapse
Affiliation(s)
- Farah Benyettou
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Mostafa Khair
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Thirumurugan Prakasam
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Sabu Varghese
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Zineb Matouk
- Technology
Innovative Institute, P.O. Box 9639, Abu Dhabi 9639, United Arab Emirates
| | - Maryam Alkaabi
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Pilar Pena-Sánchez
- Instituto
de Ciencia de Materiales de Madrid-CSIC, C. Sor Juana Inés de La Cruz 3, Madrid 28049, Spain
| | - Maylis Boitet
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Rasha AbdulHalim
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Sudhir Kumar Sharma
- Engineering
Division, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Rose Ghemrawi
- College
of Pharmacy, Al Ain University, P.O. Box 112612, Abu Dhabi 112612, United Arab Emirates
- AAU
Health and Biomedical Research Center, Al
Ain University, P.O. Box 112612, Abu Dhabi 112612, United Arab Emirates
| | - Sneha Thomas
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Jamie Whelan
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| | - Renu Pasricha
- Core Technology
Platforms, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Ramesh Jagannathan
- Engineering
Division, New York University Abu Dhabi, 129188 Abu Dhabi, United Arab Emirates
| | - Felipe Gándara
- Instituto
de Ciencia de Materiales de Madrid-CSIC, C. Sor Juana Inés de La Cruz 3, Madrid 28049, Spain
| | - Ali Trabolsi
- Chemistry
Program, New York University Abu Dhabi (NYUAD), Abu Dhabi 129188, United Arab Emirates
| |
Collapse
|
15
|
Vasileva O, Zaborova O, Shmykov B, Ivanov R, Reshetnikov V. Composition of lipid nanoparticles for targeted delivery: application to mRNA therapeutics. Front Pharmacol 2024; 15:1466337. [PMID: 39508050 PMCID: PMC11537937 DOI: 10.3389/fphar.2024.1466337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/10/2024] [Indexed: 11/08/2024] Open
Abstract
Today, lipid nanoparticles (LNPs) are some of the main delivery systems for mRNA-based therapeutics. The scope of LNP applications in terms of RNA is not limited to antiviral vaccines but encompasses anticancer drugs and therapeutics for genetic (including rare) diseases. Such widespread use implies high customizability of targeted delivery of LNPs to specific organs and tissues. This review addresses vector-free options for targeted delivery of LNPs, namely the influence of lipid composition of these nanoparticles on their biodistribution. In the review, experimental studies are examined that are focused on the biodistribution of mRNA or of the encoded protein after mRNA administration via LNPs in mammals. We also performed a comprehensive analysis of individual lipids' functional groups that ensure biodistribution to desired organs. These data will allow us to outline prospects for further optimization of lipid compositions of nanoparticles for targeted delivery of mRNA therapeutics.
Collapse
Affiliation(s)
- Olga Vasileva
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Olga Zaborova
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
- Chemistry Department, Moscow State University, Moscow, Russia
| | - Bogdan Shmykov
- Chemistry Department, Moscow State University, Moscow, Russia
| | - Roman Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Vasiliy Reshetnikov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
16
|
Selc M, Macova R, Babelova A. Novel Strategies Enhancing Bioavailability and Therapeutical Potential of Silibinin for Treatment of Liver Disorders. Drug Des Devel Ther 2024; 18:4629-4659. [PMID: 39444787 PMCID: PMC11498047 DOI: 10.2147/dddt.s483140] [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: 08/07/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Silibinin, a bioactive component found in milk thistle extract (Silybum marianum), is known to have significant therapeutic potential in the treatment of various liver diseases. It is considered a key element of silymarin, which is traditionally used to support liver function. The main mechanisms of action of silibinin are attributed to its antioxidant properties protecting liver cells from damage caused by free radicals. Experimental studies conducted in vitro and in vivo have confirmed its ability to inhibit inflammatory and fibrotic processes, as well as promote the regeneration of damaged liver tissue. Therefore, silibinin represents a promising tool for the treatment of liver diseases. Since the silibinin molecule is insoluble in water and has poor bioavailability in vivo, new perspectives on solving this problem are being sought. The two most promising approaches are the water-soluble derivative silibinin-C-2',3-dihydrogen succinate, disodium salt, and the silibinin-phosphatidylcholine complex. Both drugs are currently under evaluation in liver disease clinical trials. Nevertheless, the mechanism underlying silibinin biological activity is still elusive and its more detailed understanding would undoubtedly increase its potential in the development of effective therapeutic strategies against liver diseases. This review is focused on the therapeutic potential of silibinin and its derivates, approaches to increase the bioavailability and the benefits in the treatment of liver diseases that have been achieved so far. The review discusses the relevant in vitro and in vivo studies that investigated the protective effects of silibinin in various forms of liver damage.
Collapse
Affiliation(s)
- Michal Selc
- Centre for Advanced Material Application, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Radka Macova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Genetics, Faculty of Natural Sciences, Comenius University Bratislava, Bratislava, Slovakia
| | - Andrea Babelova
- Centre for Advanced Material Application, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
17
|
Yang T, Zhang N, Liu Y, Yang R, Wei Z, Liu F, Song D, Wang L, Wei J, Li Y, Shen D, Liang G. Nanoplatelets modified with RVG for targeted delivery of miR-375 and temozolomide to enhance gliomas therapy. J Nanobiotechnology 2024; 22:623. [PMID: 39402578 PMCID: PMC11476726 DOI: 10.1186/s12951-024-02895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Gliomas are one of the most frequent primary brain tumors and pose a serious threat to people's lives and health. Platelets, a crucial component of blood, have been applied as drug delivery carriers for disease diagnosis and treatment. In this study, we designed engineered nanoplatelets for targeted delivery of therapeutic miR-375 and temozolomide (TMZ, a first-line glioma treatment agent) to enhance glioma therapy. Nanoplatelets were prepared through mild ultrasound, TMZ and miR-375 were co-loaded through ultrasound and electrostatic interactions, respectively, to combine chemotherapy with gene therapy against glioma. To improve the blood brain barrier (BBB) crossing efficiency and glioma targeting ability, the nanoplatelets were modified with central nervous system-specific rabies viral glycoprotein peptide (RVG) through thiol-maleimide click reaction. The RVG modified nanoplatelets co-loaded TMZ and miR-375 (NR/TMZ/miR-375) not only inherited the good stability and remarkable biocompatibility of platelets, but also promoted the cellular uptake and penetration of glioma tissues, and effectively induced cell apoptosis to enhance the therapeutic effect of drugs. In vivo studies showed that NR/TMZ/miR-375 significantly increased the circulation time of TMZ, and exhibited superior combined antitumor effects. In summary, this multifunctional 'natural' nanodrug delivery system provides a potent, scalable, and safety approach for platelet-based combined cancer chemotherapy and gene therapy.
Collapse
Affiliation(s)
- Tingting Yang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
- Zhumadian Cental Hospital, Zhumadian, 463000, China
| | - Nan Zhang
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Yuanyuan Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Ruyue Yang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Zhaoyi Wei
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Futai Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Dan Song
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Longwei Wang
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Jiangyan Wei
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Yuanpei Li
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Deliang Shen
- Key Laboratory of Cardiac Injury and Repair of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Gaofeng Liang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China.
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China.
| |
Collapse
|
18
|
Andreani T, Cheng R, Elbadri K, Ferro C, Menezes T, Dos Santos MR, Pereira CM, Santos HA. Natural compounds-based nanomedicines for cancer treatment: Future directions and challenges. Drug Deliv Transl Res 2024; 14:2845-2916. [PMID: 39003425 PMCID: PMC11385056 DOI: 10.1007/s13346-024-01649-z] [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] [Accepted: 06/05/2024] [Indexed: 07/15/2024]
Abstract
Several efforts have been extensively accomplished for the amelioration of the cancer treatments using different types of new drugs and less invasives therapies in comparison with the traditional therapeutic modalities, which are widely associated with numerous drawbacks, such as drug resistance, non-selectivity and high costs, restraining their clinical response. The application of natural compounds for the prevention and treatment of different cancer cells has attracted significant attention from the pharmaceuticals and scientific communities over the past decades. Although the use of nanotechnology in cancer therapy is still in the preliminary stages, the application of nanotherapeutics has demonstrated to decrease the various limitations related to the use of natural compounds, such as physical/chemical instability, poor aqueous solubility, and low bioavailability. Despite the nanotechnology has emerged as a promise to improve the bioavailability of the natural compounds, there are still limited clinical trials performed for their application with various challenges required for the pre-clinical and clinical trials, such as production at an industrial level, assurance of nanotherapeutics long-term stability, physiological barriers and safety and regulatory issues. This review highlights the most recent advances in the nanocarriers for natural compounds secreted from plants, bacteria, fungi, and marine organisms, as well as their role on cell signaling pathways for anticancer treatments. Additionally, the clinical status and the main challenges regarding the natural compounds loaded in nanocarriers for clinical applications were also discussed.
Collapse
Affiliation(s)
- Tatiana Andreani
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
- GreenUPorto-Sustainable Agrifood Production Research Centre & Inov4Agro, Department of Biology, Faculty of Sciences of University of Porto, Rua Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Ruoyu Cheng
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute Groningen (PRECISION), University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands
| | - Khalil Elbadri
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Claudio Ferro
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Research Institute for Medicines, iMed.Ulisboa, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisbon, Portugal
| | - Thacilla Menezes
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Mayara R Dos Santos
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Carlos M Pereira
- Chemistry Research Centre (CIQUP) and Institute of Molecular Sciences (IMS), Department of Chemistry and Biochemistry, Faculty of Sciences of University of Porto, Rua Do Campo Alegre s/n, 4169-007, Porto, Portugal
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland.
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute Groningen (PRECISION), University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
19
|
Ren M, Abdullah SW, Pei C, Guo H, Sun S. Use of virus-like particles and nanoparticle-based vaccines for combating picornavirus infections. Vet Res 2024; 55:128. [PMID: 39350170 PMCID: PMC11443892 DOI: 10.1186/s13567-024-01383-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
Picornaviridae are non-enveloped ssRNA viruses that cause diseases such as poliomyelitis, hand-foot-and-mouth disease (HFMD), hepatitis A, encephalitis, myocarditis, and foot-and-mouth disease (FMD). Virus-like particles (VLPs) vaccines mainly comprise particles formed through the self-assembly of viral capsid proteins (for enveloped viruses, envelope proteins are also an option). They do not contain the viral genome. On the other hand, the nanoparticles vaccine (NPs) is mainly composed of self-assembling biological proteins or nanomaterials, with viral antigens displayed on the surface. The presentation of viral antigens on these particles in a repetitive array can elicit a strong immune response in animals. VLPs and NPs can be powerful platforms for multivalent antigen presentation. This review summarises the development of virus-like particle vaccines (VLPs) and nanoparticle vaccines (NPs) against picornaviruses. By detailing the progress made in the fight against various picornaviruses such as poliovirus (PV), foot-and-mouth disease virus (FMDV), enterovirus (EV), Senecavirus A (SVA), and encephalomyocarditis virus (EMCV), we in turn highlight the significant strides made in vaccine technology. These advancements include diverse construction methods, expression systems, elicited immune responses, and the use of various adjuvants. We see promising prospects for the continued development and optimisation of VLPs and NPs vaccines. Future research should focus on enhancing these vaccines' immunogenicity, stability, and delivery methods. Moreover, expanding our understanding of the interplay between these vaccines and the immune system will be crucial. We hope these insights will inspire and guide fellow researchers in the ongoing quest to combat picornavirus infections more effectively.
Collapse
Affiliation(s)
- Mei Ren
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gembloux Agro-Biotech, University of Liege, Gembloux, Belgium
| | - Sahibzada Waheed Abdullah
- Livestock and dairy development department peshawar, Government of Khyber Pakhtunkhwa, Peshawar, Pakistan
| | - Chenchen Pei
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
| |
Collapse
|
20
|
Desai DA, Schmidt S, Cristofoletti R. A quantitative systems pharmacology (QSP) platform for preclinical to clinical translation of in-vivo CRISPR-Cas therapy. Front Pharmacol 2024; 15:1454785. [PMID: 39372210 PMCID: PMC11449743 DOI: 10.3389/fphar.2024.1454785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/30/2024] [Indexed: 10/08/2024] Open
Abstract
Background: In-vivo CRISPR Cas genome editing is a complex therapy involving lipid nanoparticle (LNP), messenger RNA (mRNA), and single guide RNA (sgRNA). This novel modality requires prior modeling to predict dose-exposure-response relationships due to limited information on sgRNA and mRNA biodistribution. This work presents a QSP model to characterize, predict, and translate the Pharmacokinetics/Pharmacodynamics (PK/PD) of CRISPR therapies from preclinical species (mouse, non-human primate (NHP)) to humans using two case studies: transthyretin amyloidosis and LDL-cholesterol reduction. Methods: PK/PD data were sourced from literature. The QSP model incorporates mechanisms post-IV injection: 1) LNP binding to opsonins in liver vasculature; 2) Phagocytosis into the Mononuclear Phagocytotic System (MPS); 3) LNP internalization via endocytosis and LDL receptor-mediated endocytosis in the liver; 4) Cellular internalization and transgene product release; 5) mRNA and sgRNA disposition via exocytosis and clathrin-mediated endocytosis; 6) Renal elimination of LNP and sgRNA; 7) Exonuclease degradation of sgRNA and mRNA; 8) mRNA translation into Cas9 and RNP complex formation for gene editing. Monte-Carlo simulations were performed for 1000 subjects and showed a reduction in serum TTR. Results: The rate of internalization in interstitial layer was 0.039 1/h in NHP and 0.007 1/h in humans. The rate of exocytosis was 6.84 1/h in mouse, 2690 1/h in NHP, and 775 1/h in humans. Pharmacodynamics were modeled using an indirect response model, estimating first-order degradation rate (0.493 1/d) and TTR reduction parameters in NHP. Discussion: The QSP model effectively characterized biodistribution and dose-exposure relationships, aiding the development of these novel therapies. The utility of platform QSP model can be paramount in facilitating the discovery and development of these novel agents.
Collapse
Affiliation(s)
- Devam A. Desai
- Center of Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, FL, United States
| | | | - Rodrigo Cristofoletti
- Center of Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, FL, United States
| |
Collapse
|
21
|
Parrot M, Cave J, Pelaez MJ, Ghandehari H, Dogra P, Yellepeddi V. A Minimal PBPK Model Describes the Differential Disposition of Silica Nanoparticles In Vivo. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.18.24313941. [PMID: 39371117 PMCID: PMC11451661 DOI: 10.1101/2024.09.18.24313941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Nanoparticles (NPs) have emerged as promising candidates for drug delivery due to their tunable physical and chemical properties. Among these, silica nanoparticles (SiNPs) are particularly valued for their biocompatibility and adaptability in applications like drug delivery and medical imaging. However, predicting SiNP biodistribution and clearance remains a significant challenge. To address this, we developed a minimal physiologically-based pharmacokinetic (mPBPK) model to simulate the systemic disposition of SiNPs, calibrated using in vivo PK data from mice. The model assesses how variations in surface charge, size, porosity, and geometry influence SiNP biodistribution across key organs, including the kidneys, lungs, liver, and spleen. A global sensitivity analysis identified the most influential parameters, with the unbound fraction and elimination rate constants for the kidneys and MPS emerging as critical determinants of SiNP clearance. Non-compartmental analysis (NCA) further revealed that aminated SiNPs exhibit high accumulation in the liver, spleen, and kidneys, while mesoporous SiNPs primarily accumulate in the lungs. Rod-shaped SiNPs showed faster clearance compared to spherical NPs. The mPBPK model was extrapolated to predict SiNP behavior in humans, yielding strong predictive accuracy with Pearson correlation coefficients of 0.98 for mice and 0.92 for humans. This model provides a robust framework for predicting the pharmacokinetics of diverse SiNPs, offering valuable insights for optimizing NP-based drug delivery systems and guiding the translation of these therapies from preclinical models to human applications.
Collapse
|
22
|
Su S, Shen X, Shi X, Li X, Chen J, Yang W, Sun M, Tang YD, Wang H, Wang S, Cai X, Lu Y, An T, Yang Y, Meng F. Cell-penetrating peptides TAT and 8R functionalize P22 virus-like particles to enhance tissue distribution and retention in vivo. Front Vet Sci 2024; 11:1460973. [PMID: 39290505 PMCID: PMC11405305 DOI: 10.3389/fvets.2024.1460973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Virus-like particles (VLPs) are used as nanocontainers for targeted drug, protein, and vaccine delivery. The phage P22 VLP is an ideal macromolecule delivery vehicle, as it has a large exterior surface area, which facilitates multivalent genetic and chemical modifications for cell recognition and penetration. Arginine-rich cell-penetrating peptides (CPPs) can increase cargo transport efficiency in vivo. However, studies on the tissue distribution and retention of P22 VLPs mediated by TAT and 8R are lacking. This study aimed to analyze the TAT and 8R effects on the P22 VLPs transport efficiency and tissue distribution both in vitro and in vivo. We used a prokaryotic system to prepare P22 VLP self-assembled particles and expressed TAT-or 8R-conjugated mCherry on the VLP capsid protein as model cargoes and revealed that the level of P22 VLP-mCherry penetrating the cell membrane was low. However, both TAT and 8R significantly promoted the cellular uptake efficiency of P22 VLPs in vitro, as well as enhanced the tissue accumulation and retention of P22 VLPs in vivo. At 24 h postinjection, TAT enhanced the tissue distribution and retention in the lung, whereas 8R could be better accumulation in brain. Thus, TAT was superior in terms of cellular uptake and tissue accumulation in the P22 VLPs delivery system. Understanding CPP biocompatibility and tissue retention will expand their potential applications in macromolecular cargo delivery.
Collapse
Affiliation(s)
- Shibo Su
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuegang Shen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xinqi Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jin Chen
- Institute of Veterinary Immunology and Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Wei Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mingxia Sun
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yan-Dong Tang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Haiwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shujie Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuehui Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Research Center for Veterinary Biopharmaceutical Technology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yu Lu
- Institute of Veterinary Immunology and Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, China
| | - Tongqing An
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yongbo Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
- Heilongjiang Research Center for Veterinary Biopharmaceutical Technology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Fandan Meng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
23
|
Kirman CR, Kent B, Bigelow J, Canady RA, Chen Q, Chou WC, Li D, Lin Z, Kumar V, Paini A, Poulin P, Sweeney LM, Hays SM. Physiologically based pharmacokinetic modeling of metal nanoparticles for risk assessment of inhalation exposures: a state-of-the-science expert panel review. Nanotoxicology 2024; 18:527-541. [PMID: 39311096 DOI: 10.1080/17435390.2024.2401430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/29/2024] [Indexed: 09/29/2024]
Abstract
A critical review of the current state-of-the-science for the physiologically based pharmacokinetic (PBPK) modeling of metal nanoparticles and their application to human health risk assessment for inhalation exposures was conducted. A systematic literature search was used to identify four model groups (defined as a primary publication along with multiple supplementary publications) subject to review. Using a recent guideline document from the Organization for Economic Cooperation and Development (OECD) for PBPK model evaluation, these model groups were critically peer-reviewed by an independent panel of experts to identify those to be considered for modeling and simulation application. Based upon the expert panel input, model confidence scores for the four model groups ranged from 30 to 41 (out of a maximum score of 50). The three highest-scoring model groups were then applied to compare predictions to a different metal nanoparticle (i.e. not specifically used to parameterize the original models) using a recently published data set for tissue burdens in rats, as well as predicting human tissue burdens expected for corresponding occupational exposures. Overall, the rat models performed reasonably well in predicting the lung but tended to overestimate systemic tissue burdens. Data needs for improving the state-of-the-science, including quantitative particle characterization in tissues, nanoparticle-corona data, long-term exposure data, interspecies extrapolation methods, and human biomonitoring/toxicokinetic data are discussed.
Collapse
Affiliation(s)
| | - B Kent
- Waterborne, Leesburg, Virginia, USA
| | | | - R A Canady
- NeutralScience, Camano Island, Washington, USA
| | - Q Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA
| | - W C Chou
- Department of Environmental Sciences, College of Natural & Agricultural Sciences, University of California, Riverside, California, USA
| | - D Li
- School of Public Health, University of Nevada, Reno, Nevada, USA
| | - Z Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA
| | - V Kumar
- Environmental Engineering Laboratory, Department d'Enginyeria Quimica, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
- IISPV, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, Reus, Spain
| | - A Paini
- esqLABS GmbH, Saterland, Germany
| | - P Poulin
- Patrick Poulin Inc, Québec City, Canada
- School of Public Health, University of Montréal, Montréal, Canada
| | | | - S M Hays
- SciPinion, Bozeman, Montana, USA
| |
Collapse
|
24
|
Longobardi G, Moore TL, Conte C, Ungaro F, Satchi‐Fainaro R, Quaglia F. Polyester nanoparticles delivering chemotherapeutics: Learning from the past and looking to the future to enhance their clinical impact in tumor therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1990. [PMID: 39217459 PMCID: PMC11670051 DOI: 10.1002/wnan.1990] [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: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Polymeric nanoparticles (NPs), specifically those comprised of biodegradable and biocompatible polyesters, have been heralded as a game-changing drug delivery platform. In fact, poly(α-hydroxy acids) such as polylactide (PLA), poly(lactide-co-glycolide) (PLGA), and poly(ε-caprolactone) (PCL) have been heavily researched in the past three decades as the material basis of polymeric NPs for drug delivery applications. As materials, these polymers have found success in resorbable sutures, biodegradable implants, and even monolithic, biodegradable platforms for sustained release of therapeutics (e.g., proteins and small molecules) and diagnostics. Few fields have gained more attention in drug delivery through polymeric NPs than cancer therapy. However, the clinical translational of polymeric nanomedicines for treating solid tumors has not been congruent with the fervor or funding in this particular field of research. Here, we attempt to provide a comprehensive snapshot of polyester NPs in the context of chemotherapeutic delivery. This includes a preliminary exploration of the polymeric nanomedicine in the cancer research space. We examine the various processes for producing polyester NPs, including methods for surface-functionalization, and related challenges. After a detailed overview of the multiple factors involved with the delivery of NPs to solid tumors, the crosstalk between particle design and interactions with biological systems is discussed. Finally, we report state-of-the-art approaches toward effective delivery of NPs to tumors, aiming at identifying new research areas and re-evaluating the reasons why some research avenues have underdelivered. We hope our effort will contribute to a better understanding of the gap to fill and delineate the future research work needed to bring polyester-based NPs closer to clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Thomas Lee Moore
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Claudia Conte
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Francesca Ungaro
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- Sagol School of NeurosciencesTel Aviv UniversityTel AvivIsrael
| | - Fabiana Quaglia
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
25
|
Choi W, Kohane DS. Hybrid Nanoparticle-Hydrogel Systems for Drug Delivery Depots and Other Biomedical Applications. ACS NANO 2024; 18:22780-22792. [PMID: 39140388 DOI: 10.1021/acsnano.4c06888] [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: 08/15/2024]
Abstract
Hydrogel-based depots typically tend to remain where injected and have excellent biocompatibility but are relatively poor at controlling drug release. Nanoparticles (NPs) typically have the opposite properties. The smaller the NPs are, the more likely they are to leave the site of injection. Their biocompatibility is variable depending on the material but can be poor. However, NPs can be good at controlling drug release. In these and other properties, combining NPs and hydrogels can leverage their advantages and negate their disadvantages. This review highlights the rationale for hybrid NP-hydrogel systems in drug delivery, the basic methods of producing them, and examples where combining the two systems addressed specific problems.
Collapse
Affiliation(s)
- Wonmin Choi
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
26
|
Mi K, Chou WC, Chen Q, Yuan L, Kamineni VN, Kuchimanchi Y, He C, Monteiro-Riviere NA, Riviere JE, Lin Z. Predicting tissue distribution and tumor delivery of nanoparticles in mice using machine learning models. J Control Release 2024; 374:219-229. [PMID: 39146980 DOI: 10.1016/j.jconrel.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/24/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
Nanoparticles (NPs) can be designed for targeted delivery in cancer nanomedicine, but the challenge is a low delivery efficiency (DE) to the tumor site. Understanding the impact of NPs' physicochemical properties on target tissue distribution and tumor DE can help improve the design of nanomedicines. Multiple machine learning and artificial intelligence models, including linear regression, support vector machine, random forest, gradient boosting, and deep neural networks (DNN), were trained and validated to predict tissue distribution and tumor delivery based on NPs' physicochemical properties and tumor therapeutic strategies with the dataset from Nano-Tumor Database. Compared to other machine learning models, the DNN model had superior predictions of DE to tumors and major tissues. The determination coefficients (R2) for the test datasets were 0.41, 0.42, 0.45, 0.79, 0.87, and 0.83 for DE in tumor, heart, liver, spleen, lung, and kidney, respectively. All the R2 and root mean squared error (RMSE) results of the test datasets were similar to the 5-fold cross validation results. Feature importance analysis showed that the core material of NPs played an important role in output predictions among all physicochemical properties. Furthermore, multiple NP formulations with greater DE to the tumor were determined by the DNN model. To facilitate model applications, the final model was converted to a web dashboard. This model could serve as a high-throughput pre-screening tool to support the design of new and efficient nanomedicines with greater tumor DE and serve as an alternative tool to reduce, refine, and partially replace animal experimentation in cancer nanomedicine research.
Collapse
Affiliation(s)
- Kun Mi
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Wei-Chun Chou
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA; Department of Environmental Sciences, College of Natural & Agricultural Sciences, University of California, Riverside, CA 92521, USA
| | - Qiran Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Long Yuan
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Venkata N Kamineni
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Yashas Kuchimanchi
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Chunla He
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA
| | - Nancy A Monteiro-Riviere
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS 66506, USA; Center for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, NC 27606, USA
| | - Jim E Riviere
- Center for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, NC 27606, USA; 1Data Consortium, Kansas State University, Olathe, KS 66061, USA
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
27
|
Palomino-Cano C, Moreno E, Irache JM, Espuelas S. Targeting and activation of macrophages in leishmaniasis. A focus on iron oxide nanoparticles. Front Immunol 2024; 15:1437430. [PMID: 39211053 PMCID: PMC11357945 DOI: 10.3389/fimmu.2024.1437430] [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: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages play a pivotal role as host cells for Leishmania parasites, displaying a notable functional adaptability ranging from the proinflammatory, leishmanicidal M1 phenotype to the anti-inflammatory, parasite-permissive M2 phenotype. While macrophages can potentially eradicate amastigotes through appropriate activation, Leishmania employs diverse strategies to thwart this activation and redirect macrophages toward an M2 phenotype, facilitating its survival and replication. Additionally, a competition for iron between the two entities exits, as iron is vital for both and is also implicated in macrophage defensive oxidative mechanisms and modulation of their phenotype. This review explores the intricate interplay between macrophages, Leishmania, and iron. We focus the attention on the potential of iron oxide nanoparticles (IONPs) as a sort of immunotherapy to treat some leishmaniasis forms by reprogramming Leishmania-permissive M2 macrophages into antimicrobial M1 macrophages. Through the specific targeting of iron in macrophages, the use of IONPs emerges as a promising strategy to finely tune the parasite-host interaction, endowing macrophages with an augmented antimicrobial arsenal capable of efficiently eliminating these intrusive microbes.
Collapse
Affiliation(s)
- Carmen Palomino-Cano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Esther Moreno
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Juan M. Irache
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| | - Socorro Espuelas
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Medical Research Institute (IdiSNA), Pamplona, Spain
| |
Collapse
|
28
|
Littrell CA, Takacs GP, Sankara CS, Sherman A, Rubach KA, Garcia JS, Bell CA, Lnu T, Harrison JK, Zhang F. Systemically targeting monocytic myloid-derrived suppressor cells using dendrimers and their cell-level biodistribution kinetics. J Control Release 2024; 374:181-193. [PMID: 39103055 DOI: 10.1016/j.jconrel.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
The focus of nanoparticles in vivo trafficking has been mostly on their tissue-level biodistribution and clearance. Recent progress in the nanomedicine field suggests that the targeting of nanoparticles to immune cells can be used to modulate the immune response and enhance therapeutic delivery to the diseased tissue. In the presence of tumor lesions, monocytic-myeloid-derived suppressor cells (M-MDSCs) expand significantly in the bone marrow, egress into peripheral blood, and traffic to the solid tumor, where they help maintain an immuno-suppressive tumor microenvironment. In this study, we investigated the interaction between PAMAM dendrimers and M-MDSCs in two murine models of glioblastoma, by examining the cell-level biodistribution kinetics of the systemically injected dendrimers. We found that M-MDSCs in the tumor and lymphoid organs can efficiently endocytose hydroxyl dendrimers. Interestingly, the trafficking of M-MDSCs from the bone marrow to the tumor contributed to the deposition of hydroxyl dendrimers in the tumor. M-MDSCs showed different capacities of endocytosing dendrimers of different functionalities in vivo. This differential uptake was mediated by the unique serum proteins associated with each dendrimer surface functionality. The results of this study set up the framework for developing dendrimer-based immunotherapy to target M-MDSCs for cancer treatment.
Collapse
Affiliation(s)
- Chad A Littrell
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Gregory P Takacs
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Chenikkayala Siva Sankara
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Alexandra Sherman
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Kai A Rubach
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Julia S Garcia
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Coral A Bell
- Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Tejashwini Lnu
- Department of Chemical Engineering, University of Florida College of Pharmacy, Gainesville, FL, United States
| | - Jeffrey K Harrison
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Fan Zhang
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States; Department of Pharmaceutics, University of Florida College of Pharmacy, Gainesville, FL, United States; Department of Chemical Engineering, University of Florida College of Pharmacy, Gainesville, FL, United States.
| |
Collapse
|
29
|
Rojekar S, Gholap AD, Togre N, Bhoj P, Haeck C, Hatvate N, Singh N, Vitore J, Dhoble S, Kashid S, Patravale V. Current status of mannose receptor-targeted drug delivery for improved anti-HIV therapy. J Control Release 2024; 372:494-521. [PMID: 38849091 DOI: 10.1016/j.jconrel.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
In the pursuit of achieving better therapeutic outcomes in the treatment of HIV, innovative drug delivery strategies have been extensively explored. Mannose receptors, which are primarily found on macrophages and dendritic cells, offer promising targets for drug delivery due to their involvement in HIV pathogenesis. This review article comprehensively evaluates recent drug delivery system advancements targeting the mannose receptor. We have systematically described recent developments in creating and utilizing drug delivery platforms, including nanoparticles, liposomes, micelles, noisomes, dendrimers, and other nanocarrier systems targeted at the mannose receptor. These strategies aim to enhance drug delivery specificity, bioavailability, and therapeutic efficacy while decreasing off-target effects and systemic toxicity. Furthermore, the article delves into how mannose receptors and HIV interact, highlighting the potential for exploiting this interaction to enhance drug delivery to infected cells. The review covers essential topics, such as the rational design of nanocarriers for mannose receptor recognition, the impact of physicochemical properties on drug delivery performance, and how targeted delivery affects the pharmacokinetics and pharmacodynamics of anti-HIV agents. The challenges of these novel strategies, including immunogenicity, stability, and scalability, and future research directions in this rapidly growing area are discussed. The knowledge synthesis presented in this review underscores the potential of mannose receptor-based targeted drug delivery as a promising avenue for advancing HIV treatment. By leveraging the unique properties of mannose receptors, researchers can design drug delivery systems that cater to individual needs, overcome existing limitations, and create more effective and patient-friendly treatments in the ongoing fight against HIV/AIDS.
Collapse
Affiliation(s)
- Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Namdev Togre
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Priyanka Bhoj
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Clement Haeck
- Population Council, , Center for Biomedical Research, 1230 York Avenue, New York, NY 10065, USA
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India
| | - Jyotsna Vitore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Sagar Dhoble
- Department of Pharmacology and Toxicology, R. K. Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Snehal Kashid
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
30
|
Zong Q, He C, Long B, Huang Q, Chen Y, Li Y, Dang Y, Cai C. Targeted Delivery of Nanoparticles to Blood Vessels for the Treatment of Atherosclerosis. Biomedicines 2024; 12:1504. [PMID: 39062077 PMCID: PMC11275173 DOI: 10.3390/biomedicines12071504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis is a common form of cardiovascular disease, which is one of the most prevalent causes of death worldwide, particularly among older individuals. Surgery is the mainstay of treatment for severe stenotic lesions, though the rate of restenosis remains relatively high. Current medication therapy for atherosclerosis has limited efficacy in reversing the formation of atherosclerotic plaques. The search for new drug treatment options is imminent. Some potent medications have shown surprising therapeutic benefits in inhibiting inflammation and endothelial proliferation in plaques. Unfortunately, their use is restricted due to notable dose-dependent systemic side effects or degradation. Nevertheless, with advances in nanotechnology, an increasing number of nano-related medical applications are emerging, such as nano-drug delivery, nano-imaging, nanorobots, and so forth, which allow for restrictions on the use of novel atherosclerotic drugs to be lifted. This paper reviews new perspectives on the targeted delivery of nanoparticles to blood vessels for the treatment of atherosclerosis in both systemic and local drug delivery. In systemic drug delivery, nanoparticles inhibit drug degradation and reduce systemic toxicity through passive and active pathways. To further enhance the precise release of drugs, the localized delivery of nanoparticles can also be accomplished through blood vessel wall injection or using endovascular interventional devices coated with nanoparticles. Overall, nanotechnology holds boundless potential for the diagnosis and treatment of atherosclerotic diseases in the future.
Collapse
Affiliation(s)
- Qiushuo Zong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chengyi He
- Department of Vascular Surgery, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Binbin Long
- Department of General Surgery, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan 442099, China;
| | - Qingyun Huang
- Department of Cardiothoracic Surgery, The First Hospital of Putian Affiliated to Fujian Medical University, Putian 351106, China;
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| |
Collapse
|
31
|
Zhou Y, Zhou XX, Jiang H, Liu W, Chen F, Gardea-Torresdey JL, Yan B. In Vitro Toxicity and Modeling Reveal Nanoplastic Effects on Marine Bivalves. ACS NANO 2024; 18:17228-17239. [PMID: 38877988 DOI: 10.1021/acsnano.4c04607] [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: 07/03/2024]
Abstract
Nanoplastics (NPs) represent a growing concern for global environmental health, particularly in marine ecosystems where they predominantly accumulate. The impact of NPs on marine benthic organisms, such as bivalves, raises critical questions regarding ecological integrity and food safety. Traditional methods for assessing NP toxicity are often limited by their time-intensive nature and ethical considerations. Herein, we explore the toxicological effects of NPs on the marine bivalve Ruditapes philippinarum, employing a combination of in vitro cellular assays and advanced modeling techniques. Results indicate a range of adverse effects at the organismal level, including growth inhibition (69.5-108%), oxidative stress, lipid peroxidation, and DNA damage in bivalves, following exposure to NPs at concentrations in the range of 1.6 × 109-1.6 × 1011 particles/mL (p/mL). Interestingly, the growth inhibition predicted by models (54.7-104%), based on in vitro cellular proliferation assays, shows strong agreement with the in vivo outcomes of NP exposure. Furthermore, we establish a clear correlation between cytotoxicity observed in vitro and the toxicological responses at the organismal level. Taken together, this work suggests that the integration of computational modeling with in vitro toxicity assays can predict the detrimental effects of NPs on bivalves, offering insightful references for assessing the environmental risk assessment of NPs in marine benthic ecosystems.
Collapse
Affiliation(s)
- Yanfei Zhou
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Xiao-Xia Zhou
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Hao Jiang
- CAS Key Laboratory of Aquatic Botany and Watershed Ecology, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Wenzhi Liu
- CAS Key Laboratory of Aquatic Botany and Watershed Ecology, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan 430074, China
| | - Fengyuan Chen
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Jorge L Gardea-Torresdey
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Bing Yan
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|
32
|
Wehn AC, Krestel E, Harapan BN, Klymchenko A, Plesnila N, Khalin I. To see or not to see: In vivo nanocarrier detection methods in the brain and their challenges. J Control Release 2024; 371:216-236. [PMID: 38810705 DOI: 10.1016/j.jconrel.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
Nanoparticles have a great potential to significantly improve the delivery of therapeutics to the brain and may also be equipped with properties to investigate brain function. The brain, being a highly complex organ shielded by selective barriers, requires its own specialized detection system. However, a significant hurdle to achieve these goals is still the identification of individual nanoparticles within the brain with sufficient cellular, subcellular, and temporal resolution. This review aims to provide a comprehensive summary of the current knowledge on detection systems for tracking nanoparticles across the blood-brain barrier and within the brain. We discuss commonly employed in vivo and ex vivo nanoparticle identification and quantification methods, as well as various imaging modalities able to detect nanoparticles in the brain. Advantages and weaknesses of these modalities as well as the biological factors that must be considered when interpreting results obtained through nanotechnologies are summarized. Finally, we critically evaluate the prevailing limitations of existing technologies and explore potential solutions.
Collapse
Affiliation(s)
- Antonia Clarissa Wehn
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Eva Krestel
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany.
| | - Biyan Nathanael Harapan
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Andrey Klymchenko
- Laboratoire de Biophotonique et Pharmacologie, CNRS UMR 7213, Université de Strasbourg, 74 route du Rhin - CS 60024, 67401 Illkirch Cedex, France.
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany.
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), 14 074 Bd Henri Becquerel, 14000 Caen, France.
| |
Collapse
|
33
|
Barik D, Pidikaka C, Porel M. Dansyl-tagged xanthate ester as a capping agent to synthesize fluorescent silver nanoparticles with binding affinity toward serum albumin. Photochem Photobiol 2024; 100:980-988. [PMID: 38419115 DOI: 10.1111/php.13927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
Developing multifunctional nanomaterials with distinct photochemical properties, such as high quantum yield, improved photostability, and good biocompatibility is critical for a wide range of biomedical applications. Motivated by this, we designed and synthesized a dansyl-tagged xanthate-based capping agent (DX) for the synthesis of fluorescent silver nanoparticles (AgNPs). The capping agent DX was characterized by 1H and 13C-NMR, LC-MS, and FT-IR. The synthesized DX-capped fluorescent AgNPs were thoroughly characterized by UV-visible spectroscopy, fluorescence spectroscopy, field emission scanning electron microscope (FE-SEM), transmission electron microscope (TEM), dynamic light scattering (DLS), and zeta potential. The fluorescent AgNPs showed distinct surface plasmon resonance absorption at λmax = 414 nm, fluorescence at λmax = 498 nm, quantum yield = 0.24, zeta potential = +18.6 mV, average size = 18.2 nm. Furthermore, the biological activity of the fluorescent AgNPs was validated by its interaction with the most abundant protein in the blood, that is, BSA (Bovine serum albumin) and HSA (Human serum albumin) with binding constant of 2.34 × 104 M-1 and 2.14 × 104 M-1 respectively. Interestingly, fluorescence resonance energy transfer (FRET) was observed between the fluorescent AgNPs and BSA/HSA with a FRET efficiency of 77.23% and 56.36%, respectively, indicating strong interaction between fluorescent AgNPs and BSA/HSA.
Collapse
Affiliation(s)
- Debashis Barik
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, India
| | | | - Mintu Porel
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, India
- Environmental Sciences and Sustainable Engineering Center, Indian Institute of Technology Palakkad, Palakkad, India
| |
Collapse
|
34
|
Silveira RF, Lima AL, Gross IP, Gelfuso GM, Gratieri T, Cunha-Filho M. The role of artificial intelligence and data science in nanoparticles development: a review. Nanomedicine (Lond) 2024; 19:1271-1283. [PMID: 38905147 PMCID: PMC11285233 DOI: 10.1080/17435889.2024.2359355] [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: 03/18/2024] [Accepted: 05/21/2024] [Indexed: 06/23/2024] Open
Abstract
Artificial intelligence has revolutionized many sectors with unparalleled predictive capabilities supported by machine learning (ML). So far, this tool has not been able to provide the same level of development in pharmaceutical nanotechnology. This review discusses the current data science methodologies related to polymeric drug-loaded nanoparticle production from an innovative multidisciplinary perspective while considering the strictest data science practices. Several methodological and data interpretation flaws were identified by analyzing the few qualified ML studies. Most issues lie in following appropriate analysis steps, such as cross-validation, balancing data, or testing alternative models. Thus, better-planned studies following the recommended data science analysis steps along with adequate numbers of experiments would change the current landscape, allowing the exploration of the full potential of ML.
Collapse
Affiliation(s)
- Rodrigo Fonseca Silveira
- Laboratory of Food, Drugs, & Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Ana Luiza Lima
- Laboratory of Food, Drugs, & Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Idejan Padilha Gross
- Laboratory of Food, Drugs, & Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Guilherme Martins Gelfuso
- Laboratory of Food, Drugs, & Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Tais Gratieri
- Laboratory of Food, Drugs, & Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Marcilio Cunha-Filho
- Laboratory of Food, Drugs, & Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| |
Collapse
|
35
|
Wang M, Chen S, Cheng S, Nederstigt TAP, Poelmann RE, DeRuiter MC, Lamers GEM, Willemse JJ, Mascitelli C, Vijver MG, Richardson MK. The biodistribution of polystyrene nanoparticles administered intravenously in the chicken embryo. ENVIRONMENT INTERNATIONAL 2024; 188:108723. [PMID: 38744045 DOI: 10.1016/j.envint.2024.108723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024]
Abstract
Nanoplastics can cause severe malformations in chicken embryos. To improve our understanding of the toxicity of nanoplastics to embryos, we have studied their biodistribution in living chicken embryos. We injected the embryos in the vitelline vein at stages 18-19. We injected polystyrene nanoparticles (PS-NPs) tagged with europium- or fluorescence. Their biodistribution was tracked using inductively-coupled plasma mass spectrometry on tissue lysates, paraffin histology, and vibratome sections analysed by machine learning algorithms. PS-NPs were found at high levels in the heart, liver and kidneys. Furthermore, PS-NPs crossed the endocardium of the heart at sites of epithelial-mesenchymal transformation; they also crossed the liver endothelium. Finally, we detected PS-NPs in the allantoic fluid, consistent with their being excreted by the kidneys. Our study shows the power of the chicken embryo model for analysing the biodistribution of nanoplastics in embryos. Such experiments are difficult or impossible in mammalian embryos. These findings are a major advance in our understanding of the biodistribution and tissue-specific accumulation of PS-NPs in developing animals.
Collapse
Affiliation(s)
- Meiru Wang
- Institute of Biology, Leiden University, Sylvius Laboratory, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Shuhao Chen
- Institute of Biology, Leiden University, Sylvius Laboratory, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Shixiong Cheng
- Institute of Biology, Leiden University, Sylvius Laboratory, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Tom A P Nederstigt
- Centrum voor Milieuwetenschappen Leiden (CML), Van Steenis Building, Einsteinweg 2, 2333 CC Leiden, The Netherlands
| | - Robert E Poelmann
- Institute of Biology, Leiden University, Sylvius Laboratory, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Marco C DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, LUMC Onderzoeksgebouw, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Gerda E M Lamers
- Institute of Biology, Leiden University, Sylvius Laboratory, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Joost J Willemse
- Institute of Biology, Leiden University, Sylvius Laboratory, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Chiara Mascitelli
- Institute of Biology, Leiden University, Sylvius Laboratory, Sylviusweg 72, 2333 BE Leiden, The Netherlands
| | - Martina G Vijver
- Centrum voor Milieuwetenschappen Leiden (CML), Van Steenis Building, Einsteinweg 2, 2333 CC Leiden, The Netherlands
| | - Michael K Richardson
- Institute of Biology, Leiden University, Sylvius Laboratory, Sylviusweg 72, 2333 BE Leiden, The Netherlands.
| |
Collapse
|
36
|
Zimmer O, Goepferich A. On the uncertainty of the correlation between nanoparticle avidity and biodistribution. Eur J Pharm Biopharm 2024; 198:114240. [PMID: 38437906 DOI: 10.1016/j.ejpb.2024.114240] [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: 11/17/2023] [Revised: 02/05/2024] [Accepted: 02/28/2024] [Indexed: 03/06/2024]
Abstract
The specific delivery of a drug to its site of action also known as targeted drug delivery is a topic in the field of pharmaceutics studied for decades. One approach extensively investigated in this context is the use ligand functionalized nanoparticles. These particles are modified to carry receptor specific ligands, enabling them to accumulate at a desired target site. However, while this concept initially appears straightforward to implement, in-depth research has revealed several challenges hindering target site specific particle accumulation - some of which remain unresolved to this day. One of these challenges consists in the still incomplete understanding of how nanoparticles interact with biological systems. This knowledge gap significantly compromises the predictability of particle distribution in biological systems, which is critical for therapeutic efficacy. One of the most crucial steps in delivery is the attachment of nanoparticles to cells at the target site. This attachment occurs via the formation of multiple ligand receptor bonds. A process also referred to as multivalent interaction. While multivalency has been described extensively for individual molecules and macromolecules respectively, little is known on the multivalent binding of nanoparticles to cells. Here, we will specifically introduce the concept of avidity as a measure for favorable particle membrane interactions. Also, an overview about nanoparticle and membrane properties affecting avidity will be given. Thereafter, we provide a thorough review on literature investigating the correlation between nanoparticle avidity and success in targeted particle delivery. In particular, we want to analyze the currently uncertain data on the existence and nature of the correlation between particle avidity and biodistribution.
Collapse
Affiliation(s)
- Oliver Zimmer
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany.
| |
Collapse
|
37
|
Marques AC, Costa PC, Velho S, Amaral MH. Rheological and Injectability Evaluation of Sterilized Poloxamer-407-Based Hydrogels Containing Docetaxel-Loaded Lipid Nanoparticles. Gels 2024; 10:307. [PMID: 38786224 PMCID: PMC11121564 DOI: 10.3390/gels10050307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Nanostructured lipid carriers (NLCs) have the potential to increase the bioavailability and reduce the side effects of docetaxel (DTX). However, only a small fraction of nanoparticles given intravenously can reach a solid tumor. In situ-forming gels combined with nanoparticles facilitate local administration and promote drug retention at the tumor site. Injectable hydrogels based on poloxamer 407 are excellent candidates for this hybrid nanoparticle-hydrogel system because of their thermoresponsive behavior and biocompatibility. Therefore, this work aimed to develop injectable poloxamer hydrogels containing NLCs for intratumoral delivery of DTX. To ensure sterility, the obtained hydrogels were autoclaved (121 °C for 15 min) after preparation. Then, the incorporation of NLCs into the poloxamer hydrogels and the impact of steam sterilization on the nanocomposite hydrogels were evaluated concerning sol-gel transition, injectability, and physicochemical stability. All formulations were extruded through the tested syringe-needle systems with acceptable force (2.2-13.4 N) and work (49.5-317.7 N·mm) of injection. Following steam sterilization, injection became easier in most cases, and the physicochemical properties of all hydrogels remained practically unchanged according to the spectroscopical and thermal analysis. The rheological evaluation revealed that the nanocomposite hydrogels were liquid at 25 °C and underwent rapid gelation at 37 °C. However, their sterilized counterparts gelled at 1-2 °C above body temperature, suggesting that the autoclaving conditions employed had rendered these nanocomposite hydrogels unsuitable for local drug delivery.
Collapse
Affiliation(s)
- Ana Camila Marques
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paulo C. Costa
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Sérgia Velho
- i3S—Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Maria Helena Amaral
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
38
|
Lu G, Li B, Lin L, Li X, Ban J. Mechanical strength affecting the penetration in microneedles and PLGA nanoparticle-assisted drug delivery: Importance of preparation and formulation. Biomed Pharmacother 2024; 173:116339. [PMID: 38428314 DOI: 10.1016/j.biopha.2024.116339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
Microneedles (MNs) prepared from polymeric materials are painless and minimally invasive, safe and efficient, but they hindered by low mechanical strength and single diverse drug release pattern. Due to the distinctive mechanical strength and dimensions of poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), the integration of nano-technology with microneedles can effectively improve penetration and delivery efficiency through the stratum corneum. We herein designed a simple paroxetine (PAX)-loaded PLGA nanoparticles-integrated dissolving microneedles system (PAX-NPs-DMNs), aiming to improve the bioavailability of PAX through the synergistic permeation-enhancing effect of dissolving microneedles (DMNs) and NPs. PAX-NPs-DMNs had a complete tips molding rate (Neff) of (94.06 ± 2.16) %, a 15×15 quadrangular-conical microneedle array and an overall fracture force of 301.10 N, which were improved nearly 0.50 times compared with the blank microneedles (HA-DMNs) and PAX microneedles (PAX-DMNs). PAX-NPs-DMNs could extend the release duration of PAX from 1 h to 24 h and the cumulative permeability per unit area (Qn) was 47.66 times and 7.37 times higher than the PAX and the PAX-DMNs groups. PAX-NPs-DMNs could be rapidly dissolved within 10 min without hindering skin healing or causing adverse reactions. This study confirmed that PAX-NPs-DMNs can effectively improve the bioavailability of PAX and the mechanical strength of DMNs, which can easily penetrate the skin to provide sustained and painless delivery without causing adverse effects, thus offering a more convenient and effective method for central nervous diseases.
Collapse
Affiliation(s)
- Geng Lu
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Baohua Li
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Luping Lin
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Xiaofang Li
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Junfeng Ban
- The Innovation Team for Integrating Pharmacy with Entrepreneurship, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China.
| |
Collapse
|
39
|
Chen CY, Lin Z. Exploring the potential and challenges of developing physiologically-based toxicokinetic models to support human health risk assessment of microplastic and nanoplastic particles. ENVIRONMENT INTERNATIONAL 2024; 186:108617. [PMID: 38599027 DOI: 10.1016/j.envint.2024.108617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/05/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024]
Abstract
Microplastics (MPs) and nanoplastics (NPs) pollution has emerged as a significant and widespread environmental issue. Humans are inevitably exposed to MPs and NPs via ingestion, inhalation, and dermal contacts from various sources. However, mechanistic knowledge of their distribution, interaction, and potency in the body is still lacking. To address this knowledge gap, we have undertaken the task of elucidating the toxicokinetic (TK) behaviors of MPs and NPs, aiming to provide mechanistic information for constructing a conceptual physiologically based toxicokinetic (PBTK) model to support in silico modeling approaches. Our effort involved a thorough examination of the existing literature and data collation on the presence of MPs in the human body and in vitro/ex vivo/in vivo biodistribution across various cells and tissues. By comprehending the absorption, distribution, metabolism, and excretion mechanisms of MPs and NPs in relation to their physicochemical attributes, we established a foundational understanding of the link between external exposure and internal tissue dosimetry. We observed that particle size and surface chemistry have been thoroughly explored in previous experimental studies. However, certain attributes, such as polymer type, shape, and biofilm/biocorona, warrant attention and further examination. We discussed the fundamental disparities in TK properties of MPs/NPs from those of engineered nanoparticles. We proposed a preliminary PBTK framework with several possible modeling approaches and discussed existing challenges for further investigation. Overall, this article provides a comprehensive compilation of existing TK data of MPs/NPs, a critical overview of TK processes and mechanisms, and proposes potential PBTK modeling approaches, particularly regarding their applicability to the human system, and outlines future perspectives for developing PBTK models and their integration into human health risk assessment of MPs and NPs.
Collapse
Affiliation(s)
- Chi-Yun Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, United States; Center for Environmental and Human Toxicology, University of Florida, FL 32608, United States
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, United States; Center for Environmental and Human Toxicology, University of Florida, FL 32608, United States.
| |
Collapse
|
40
|
Wang X, Wu J, Ye H, Zhao X, Zhu S. Research Landscape of Physiologically Based Pharmacokinetic Model Utilization in Different Fields: A Bibliometric Analysis (1999-2023). Pharm Res 2024; 41:609-622. [PMID: 38383936 DOI: 10.1007/s11095-024-03676-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/05/2024] [Indexed: 02/23/2024]
Abstract
PURPOSE The physiologically based pharmacokinetic (PBPK) modeling has received increasing attention owing to its excellent predictive abilities. However, there has been no bibliometric analysis about PBPK modeling. This research aimed to summarize the research development and hot points in PBPK model utilization overall through bibliometric analysis. METHODS We searched for publications related to the PBPK modeling from 1999 to 2023 in the Web of Science Core Collection (WoSCC) database. The Microsoft Office Excel, CiteSpace and VOSviewers were used to perform the analyses. RESULTS A total of 4,649 records from 1999 to 2023 were identified, and the largest number of publications focused in the period 2018-2023. The United States was the leading country, and the Environmental Protection Agency (EPA) was the leading institution. The journal Drug Metabolism and Disposition published and co-cited the most articles. Drug-drug interactions, special populations, and new drug development are the main topics in this research field. CONCLUSION We first visualize the research landscape and hotspots of the PBPK modeling through bibliometric methods. Our study provides a better understanding for researchers, especially beginners about the dynamization of PBPK modeling and presents the relevant trend in the future.
Collapse
Affiliation(s)
- Xin Wang
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Jiangfan Wu
- School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Hongjiang Ye
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaofang Zhao
- School of Pharmacy, Chongqing Medical University, Chongqing, China
- Qiandongnan Miao and Dong Autonomous Prefecture People's Hospital, Guizhou, 556000, China
| | - Shenyin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
41
|
Shen H, Pan L, Ning K, Fang Y, Muhitdinov B, Liu E, Huang Y. Asiatic acid cyclodextrin inclusion micro-cocrystal for insoluble drug delivery and acute lung injury therapy enhancement. J Nanobiotechnology 2024; 22:119. [PMID: 38494523 PMCID: PMC10946140 DOI: 10.1186/s12951-024-02387-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a fatal respiratory disease caused by overreactive immune reactions (e.g., SARS-CoV-2 infection), with a high mortality rate. Its treatment is often compromised by inefficient drug delivery barriers and insufficient potency of the currently used drugs. Therefore, developing a highly effective lung-targeted drug delivery strategy is a pressing clinical need. RESULTS In this study, the micro-sized inclusion cocrystal of asiatic acid/γ-cyclodextrin (AA/γCD, with a stoichiometry molar ratio of 2:3 and a mean size of 1.8 μm) was prepared for ALI treatment. The dissolution behavior of the AA/γCD inclusion cocrystals followed a "spring-and-hover" model, which meaned that AA/γCD could dissolve from the cocrystal in an inclusion complex form, thereby promoting a significantly improved water solubility (nine times higher than free AA). This made the cyclodextrin-based inclusion cocrystals an effective solid form for enhanced drug absorption and delivery efficiency. The biodistribution experiments demonstrated AA/γCD accumulated predominantly in the lung (Cmax = 50 µg/g) after systemic administration due to the micron size-mediated passive targeting effect. The AA/γCD group showed an enhanced anti-inflammatory therapeutic effect, as evidenced by reduced levels of pro-inflammatory cytokines in the lung and bronchoalveolar lavage fluids (BALF). Histological examination confirmed that AA/γCD effectively inhibited inflammation reactions. CONCLUSION The micro-sized inclusion cocrystals AA/γCD were successfully delivered into the lungs by pulmonary administration and had a significant therapeutic effect on ALI.
Collapse
Affiliation(s)
- Huan Shen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li Pan
- School of Pharmacy, Zunyi Medical University, Zunyi, 563003, China
| | - Keke Ning
- School of Pharmacy, Zunyi Medical University, Zunyi, 563003, China
| | - Yuefei Fang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Bahtiyor Muhitdinov
- Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, 83 M. Ulughbek Street, Tashkent, 100125, Uzbekistan
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yongzhuo Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, Zunyi Medical University, Zunyi, 563003, China.
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai, 201203, China.
| |
Collapse
|
42
|
Peng Y, Yang Z, Sun H, Li J, Lan X, Liu S. Nanomaterials in Medicine: Understanding Cellular Uptake, Localization, and Retention for Enhanced Disease Diagnosis and Therapy. Aging Dis 2024:AD.2024.0206-1. [PMID: 38421835 DOI: 10.14336/ad.2024.0206-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Nanomaterials (NMs) have emerged as promising tools for disease diagnosis and therapy due to their unique physicochemical properties. To maximize the effectiveness and design of NMs-based medical applications, it is essential to comprehend the complex mechanisms of cellular uptake, subcellular localization, and cellular retention. This review illuminates the various pathways that NMs take to get from the extracellular environment to certain intracellular compartments by investigating the various mechanisms that underlie their interaction with cells. The cellular uptake of NMs involves complex interactions with cell membranes, encompassing endocytosis, phagocytosis, and other active transport mechanisms. Unique uptake patterns across cell types highlight the necessity for customized NMs designs. After internalization, NMs move through a variety of intracellular routes that affect where they are located subcellularly. Understanding these pathways is pivotal for enhancing the targeted delivery of therapeutic agents and imaging probes. Furthermore, the cellular retention of NMs plays a critical role in sustained therapeutic efficacy and long-term imaging capabilities. Factors influencing cellular retention include nanoparticle size, surface chemistry, and the cellular microenvironment. Strategies for prolonging cellular retention are discussed, including surface modifications and encapsulation techniques. In conclusion, a comprehensive understanding of the mechanisms governing cellular uptake, subcellular localization, and cellular retention of NMs is essential for advancing their application in disease diagnosis and therapy. This review provides insights into the intricate interplay between NMs and biological systems, offering a foundation for the rational design of next-generation nanomedicines.
Collapse
Affiliation(s)
- Yue Peng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhengshuang Yang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Hui Sun
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiuwan Lan
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
43
|
Chiang MC, Yang YP, Nicol CJB, Wang CJ. Gold Nanoparticles in Neurological Diseases: A Review of Neuroprotection. Int J Mol Sci 2024; 25:2360. [PMID: 38397037 PMCID: PMC10888679 DOI: 10.3390/ijms25042360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
This review explores the diverse applications of gold nanoparticles (AuNPs) in neurological diseases, with a specific focus on Alzheimer's disease (AD), Parkinson's disease (PD), and stroke. The introduction highlights the pivotal role of neuroinflammation in these disorders and introduces the unique properties of AuNPs. The review's core examines the mechanisms by which AuNPs exert neuroprotection and anti-neuro-inflammatory effects, elucidating various pathways through which they manifest these properties. The potential therapeutic applications of AuNPs in AD are discussed, shedding light on promising avenues for therapy. This review also explores the prospects of utilizing AuNPs in PD interventions, presenting a hopeful outlook for future treatments. Additionally, the review delves into the potential of AuNPs in providing neuroprotection after strokes, emphasizing their significance in mitigating cerebrovascular accidents' aftermath. Experimental findings from cellular and animal models are consolidated to provide a comprehensive overview of AuNPs' effectiveness, offering insights into their impact at both the cellular and in vivo levels. This review enhances our understanding of AuNPs' applications in neurological diseases and lays the groundwork for innovative therapeutic strategies in neurology.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Christopher J. B. Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, Cancer Biology and Genetics Division, Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Chieh-Ju Wang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
44
|
Al-Madani H, Yang Y, Refat M, He Q, Peng H, Wu A, Yang F. Quantification and biological evaluation of Zn xFe 3-xO 4 nanoparticle stiffness in a drug delivery system of MCF-7 cancer cells. J Mater Chem B 2024; 12:1636-1651. [PMID: 38270595 DOI: 10.1039/d3tb02723f] [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: 01/26/2024]
Abstract
The delivery of nanoparticles (NPs) to tumors remains challenging despite significant advancements in drug delivery technologies. Addressing this issue requires the establishment of quantitative and reliable criteria to evaluate the cellular absorption of NPs. The mechanical characteristics of NPs and their interaction with cells play a crucial role in cellular drug delivery by influencing cellular internalization. In particular, NPs' stiffness has emerged as a key factor affecting cellular uptake and viability. In this study, we synthesized ZnxFe3-xO4 NPs with varying Zn doping concentrations and conducted an extensive measurement process to investigate the impact of NP stiffness on cellular uptake and the viability of cancerous cells. Initially, the stiffness of the NPs was measured using two methods: single-molecule force spectrometry of atomic force microscopy (SMFS-AFM) and cation distribution as chemical structure analysis. The influence of NP stiffness on intracellular behavior was examined by assessing cellular uptake and viability at different time points during the incubation period. The results obtained from both stiffness measurement methods exhibited consistent trends. NPs with higher stiffness exhibited enhanced cellular uptake but exhibited reduced cellular viability compared to the lower-stiffness NPs. Our findings provide valuable insights into the influence of Zn doping concentration on the mechanical properties of ZnxFe3-xO4 NPs and their consequential impacts on cellular internalization. This study contributes to an improved comprehension of the mechanisms underlying cellular uptake and facilitates advancements in the field of drug transport, thereby enhancing the efficiency of NP-based drug delivery.
Collapse
Affiliation(s)
- Hamzah Al-Madani
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yiqian Yang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Moath Refat
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Qingxin He
- Guangxi Vocational & Technical Institute of Industry, Guangxi 530001, P. R. China
| | - Hao Peng
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China.
| | - Fang Yang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo 315201, P. R. China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516000, P. R. China.
| |
Collapse
|
45
|
Tang W, Zhang X, Hong H, Chen J, Zhao Q, Wu F. Computational Nanotoxicology Models for Environmental Risk Assessment of Engineered Nanomaterials. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:155. [PMID: 38251120 PMCID: PMC10819018 DOI: 10.3390/nano14020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/08/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Although engineered nanomaterials (ENMs) have tremendous potential to generate technological benefits in numerous sectors, uncertainty on the risks of ENMs for human health and the environment may impede the advancement of novel materials. Traditionally, the risks of ENMs can be evaluated by experimental methods such as environmental field monitoring and animal-based toxicity testing. However, it is time-consuming, expensive, and impractical to evaluate the risk of the increasingly large number of ENMs with the experimental methods. On the contrary, with the advancement of artificial intelligence and machine learning, in silico methods have recently received more attention in the risk assessment of ENMs. This review discusses the key progress of computational nanotoxicology models for assessing the risks of ENMs, including material flow analysis models, multimedia environmental models, physiologically based toxicokinetics models, quantitative nanostructure-activity relationships, and meta-analysis. Several challenges are identified and a perspective is provided regarding how the challenges can be addressed.
Collapse
Affiliation(s)
- Weihao Tang
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Xuejiao Zhang
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| | - Huixiao Hong
- National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd., Jefferson, AR 72079, USA
| | - Jingwen Chen
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian Key Laboratory on Chemicals Risk Control and Pollution Prevention Technology, School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Qing Zhao
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| |
Collapse
|
46
|
Liu S, Chowdhury EA, Xu V, Jerez A, Mahmood L, Ly BQ, Le HK, Nguyen A, Rajwade A, Meno-Tetang G, Shah DK. Whole-Body Disposition and Physiologically Based Pharmacokinetic Modeling of Adeno-Associated Viruses and the Transgene Product. J Pharm Sci 2024; 113:141-157. [PMID: 37805073 DOI: 10.1016/j.xphs.2023.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
To facilitate model-informed drug development (MIDD) of adeno-associated virus (AAV) therapy, here we have developed a physiologically based pharmacokinetic (PBPK) model for AAVs following preclinical investigation in mice. After 2E11 Vg/mouse dose of AAV8 and AAV9 encoding a monoclonal antibody (mAb) gene, whole-body disposition of both the vector and the transgene mAb was evaluated over 3 weeks. At steady-state, the following tissue-to-blood (T/B) concentration ratios were found for AAV8/9: ∼50 for liver; ∼10 for heart and muscle; ∼2 for brain, lung, kidney, adipose, and spleen; ≤1 for bone, skin, and pancreas. T/B values for mAb were compared with the antibody biodistribution coefficients, and five different clusters of organs were identified based on their transgene expression profile. All the biodistribution data were used to develop a novel AAV PBPK model that incorporates: (i) whole-body distribution of the vector; (ii) binding, internalization, and intracellular processing of the vector; (iii) transgene expression and secretion; and (iv) whole-body disposition of the secreted transgene product. The model was able to capture systemic and tissue PK of the vector and the transgene-produced mAb reasonably well. Pathway analysis of the PBPK model suggested that liver, muscle, and heart are the main contributors for the secreted transgene mAb. Unprecedented PK data and the novel PBPK model developed here provide the foundation for quantitative systems pharmacology (QSP) investigations of AAV-mediated gene therapies. The PBPK model can also serve as a quantitative tool for preclinical study design and preclinical-to-clinical translation of AAV-based gene therapies.
Collapse
Affiliation(s)
- Shufang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Ekram Ahmed Chowdhury
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Vivian Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Anthony Jerez
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Leeha Mahmood
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Bao Quoc Ly
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Huyen Khanh Le
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Anne Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Aneesh Rajwade
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| | - Guy Meno-Tetang
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
47
|
Blanco-Cabra N, Alcàcer-Almansa J, Admella J, Arévalo-Jaimes BV, Torrents E. Nanomedicine against biofilm infections: A roadmap of challenges and limitations. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1944. [PMID: 38403876 DOI: 10.1002/wnan.1944] [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: 10/18/2023] [Revised: 11/28/2023] [Accepted: 01/27/2024] [Indexed: 02/27/2024]
Abstract
Microbial biofilms are complex three-dimensional structures where sessile microbes are embedded in a polymeric extracellular matrix. Their resistance toward the host immune system as well as to a diverse range of antimicrobial treatments poses a serious health and development threat, being in the top 10 global public health threats declared by the World Health Organization. In an effort to combat biofilm-related microbial infections, several strategies have been developed to independently eliminate biofilms or to complement conventional antibiotic therapies. However, their limitations leave room for other treatment alternatives, where the application of nanotechnology to biofilm eradication has gained significant relevance in recent years. Their small size, penetration efficiency, and the design flexibility that they present makes them a promising alternative for biofilm infection treatment, although they also present set-backs. This review aims to describe the main possibilities and limitations of nanomedicine against biofilms, while covering the main aspects of biofilm formation and study, and the current therapies for biofilm treatment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Núria Blanco-Cabra
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Júlia Alcàcer-Almansa
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Joana Admella
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Betsy Verónica Arévalo-Jaimes
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapy Group (BIAT), Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
48
|
Shi C, Jian C, Wang L, Gao C, Yang T, Fu Z, Wu T. Dendritic cell hybrid nanovaccine for mild heat inspired cancer immunotherapy. J Nanobiotechnology 2023; 21:347. [PMID: 37752555 PMCID: PMC10521411 DOI: 10.1186/s12951-023-02106-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Cancer therapeutic vaccine can induce antigen-specific immune response, which has shown great potential in cancer immunotherapy. As the key factor of vaccine, antigen plays a central role in eliciting antitumor immunity. However, the insufficient antigen delivery and low efficiency of antigen presentation by dendritic cells (DCs) have greatly restricted the therapeutic efficiency of vaccine. Here we developed a kind of DC hybrid zinc phosphate nanoparticles to co-deliver antigenic peptide and photosensitive melanin. Owing to the chelating ability of Zn2+, the nanoparticles can co-encapsulate antigenic peptide and melanin with high efficiency. The nanovaccine showed good physiological stability with the hydration particle size was approximately 30 nm, and zeta potential was around - 10 mV. The nanovaccine showed homologous targeting effect to DCs in vivo and in vitro, efficiently delivering antigen to DCs. Meanwhile, the nanovaccine could effectively reflux to the tumor-draining lymph nodes. When combined with near-infrared irradiation, the nanovaccine induced effective mild heat in vitro and in vivo to promote antigen presentation. After administrating to MC38 tumor-bearing mice, the hybrid nanovaccine effectively promoted the maturation of DCs, the expansion of cytotoxic T lymphocytes and helper T cells, and the secretion of immunostimulatory cytokines, thereby significantly inhibiting tumor growth.
Collapse
Affiliation(s)
- Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Chen Jian
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lulu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chen Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ting Yang
- Affiliated Hospital of Yunnan University, Kunming, 650000, China
| | - Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Tingting Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China.
| |
Collapse
|
49
|
Li Y, Liang X, Shen C, Deng K, Zeng Z, Guo B, Xu X. Bio-Responsive Macromolecular Drug and Small-Molecular Drug Conjugates: Nanoparticulate Prodrugs for Tumor Microenvironment Heterogeneity Management and Therapeutic Response Enhancement. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301656. [PMID: 37144435 DOI: 10.1002/smll.202301656] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/07/2023] [Indexed: 05/06/2023]
Abstract
How to break through the poor response of current drug therapy, which often resulted from tumor microenvironment heterogeneity (TMH), remains an enormous challenge in the treatment of critical diseases. In this work, a practical solution on bio-responsive dual-drug conjugates for overcoming TMH and improving antitumor treatment, which integrates the advantages of macromolecular drugs and small-molecular drugs, is proposed. Nanoparticulate prodrugs based on small-molecular drug and macromolecular drug conjugates are designed as a robust weapon for programmable multidrug delivery at tumor-specific sites: the tumor microenvironment acid condition triggers delivery of macromolecular aptamer drugs (AX102) to manage TMH (including tumor stroma matrix, interstitial fluid pressure, vasculature network, blood perfusion, and oxygen distribution), and intracellular lysosomal acid condition activates rapid release of small-molecular drugs (doxorubicin and dactolisib) to enhance curative effects. As compared with doxorubicin chemotherapy, the tumor growth inhibition rate is enhanced by 47.94% after multiple tumor heterogeneity management. This work verifies that the nanoparticulate prodrugs facilitate TMH management and therapeutic response enhancements, as well as elucidates synergetic mechanisms for drug resistance reversal and metastasis inhibition. It is hoped that the nanoparticulate prodrugs will be an excellent demonstration of the co-delivery of small-molecular drugs and macromolecular drugs.
Collapse
Affiliation(s)
- Yachao Li
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Changsha, Hunan, 410082, China
| | - Xiaoyu Liang
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Cheng Shen
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Kefurong Deng
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Zenan Zeng
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Beiling Guo
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Xianghui Xu
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Changsha, Hunan, 410082, China
| |
Collapse
|
50
|
Wickline SA, Hou KK, Pan H. Peptide-Based Nanoparticles for Systemic Extrahepatic Delivery of Therapeutic Nucleotides. Int J Mol Sci 2023; 24:ijms24119455. [PMID: 37298407 DOI: 10.3390/ijms24119455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Peptide-based nanoparticles (PBN) for nucleotide complexation and targeting of extrahepatic diseases are gaining recognition as potent pharmaceutical vehicles for fine-tuned control of protein production (up- and/or down-regulation) and for gene delivery. Herein, we review the principles and mechanisms underpinning self-assembled formation of PBN, cellular uptake, endosomal release, and delivery to extrahepatic disease sites after systemic administration. Selected examples of PBN that have demonstrated recent proof of concept in disease models in vivo are summarized to offer the reader a comparative view of the field and the possibilities for clinical application.
Collapse
Affiliation(s)
- Samuel A Wickline
- Division of Cardiology, Department of Medical Engineering, University of South Florida, Tampa, FL 33602, USA
| | - Kirk K Hou
- Department of Ophthalmology, Stein and Doheny Eye Institutes, University of California, Los Angeles, CA 90095, USA
| | - Hua Pan
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|