1
|
Wang H, Li W, Chen J, Chen R, Qi Y, Shen L, Chen K, Dai L, Sheng Y, Wang A, Wang H, Chen C, Cheng X, Gu M. Delivery of small interfering RNA through lyophilized natural lipid nanoparticles: effects of natural lipid selection. PHARMACEUTICAL BIOLOGY 2025; 63:343-356. [PMID: 40314193 PMCID: PMC12051533 DOI: 10.1080/13880209.2025.2498169] [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: 10/03/2024] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
CONTEXT Lipid nanoparticles (LNPs) are the primary non-viral vectors for siRNA delivery. However, synthetic lipids face issues, such as low lysosomal escape efficiency and high cost. OBJECTIVE This study aimed to use three natural lipids to construct LNPs, optimize their preparation and freeze-drying processes, and evaluate their siRNA delivery efficiency in vitro. MATERIALS AND METHODS Coix seed lipid [Coix lacryma-jobi L. var. mayuen (Roman.) Stapf (Poaceae), CSL], Brucea javanica seed lipid [Brucea javanica (L.) Merr. (Simaroubaceae), BJL], and Soybean oil [Glycine max (L.) Merr. (Fabaceae), SO] were used to construct LNPs. The Z-average size, zeta potential, Polymer Dispersity Index, and N/P ratio of the LNPs were characterized. Transmission electron microscope was used for morphology observation and the MTS assay for cytotoxicity. Confocal laser scanning microscope assessed cell uptake, lysosomal escape, and co-localization of lipid droplets. The efficiency of siRNA knockdown was evaluated in three cells using qPCR and Western blot. The freeze-drying processes were optimized. RESULTS The optimal LNPs exhibited a size of 160-180 nm, zeta of 44-50 mV, and PDI of <0.2. At 200 μg/mL, the LNPs did not affect cell viability. CSL-LNPs, BJL-LNPs, and SO-LNPs reduced KRASG12D mRNA levels in AsPC-1 cells by 67.87 ± 3.89, 47.18 ± 7.65, and 42.52 ± 8.90%, respectively. Freeze-dried LNPs retained their basic physical properties and the three LNPs reducing KRASG12D mRNA levels by 58.47 ± 4.00, 51.83 ± 4.57, and 38.00 ± 4.89%, respectively. DISCUSSION AND CONCLUSION Natural lipids are promising components for LNPs construction, offering new avenues for siRNA delivery in gene therapy.
Collapse
Affiliation(s)
- Hangjie Wang
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Wei Li
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Junyan Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Rong Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Yuwei Qi
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Linshuang Shen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Kaidi Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Lewei Dai
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Yuxin Sheng
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - An Wang
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Hong Wang
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Chujian Chen
- Huzhou Institute for Food and Drug Control, Huzhou, P. R. China
| | - Xiao Cheng
- Huzhou Institute for Food and Drug Control, Huzhou, P. R. China
| | - Mancang Gu
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, P. R. China
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| |
Collapse
|
2
|
Zhang J, Zhang H, Yao C, Gu L, Dong S, Wu Y, Miao L. Global research hotspots and trends in DNA vaccine research: A bibliometric and visualization study from 2014 to 2024. Hum Vaccin Immunother 2025; 21:2457189. [PMID: 39871452 PMCID: PMC11776459 DOI: 10.1080/21645515.2025.2457189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/30/2024] [Accepted: 01/18/2025] [Indexed: 01/29/2025] Open
Abstract
This bibliometric and visualization study provides a comprehensive analysis of global research hotspots and trends in DNA vaccine research from 2014 to 2024. By employing data sourced from the Web of Science Core Collection, we identified a total of 3,600 articles. Our analysis reveals a declining trend in annual publications. Active countries, institutions, journals, and authors were identified, with China, the Pasteur Network, the Vaccine Journal, and David B Weiner being the most prolific contributors. Keywords cluster analysis distinguished four major research directions: infectious disease and immunity, viral challenge and vaccine development, optimization of DNA vaccine delivery systems, and cancer and immunotherapy research. The literature co-citation analysis revealed four major research hotspots, including DNA vaccines for Zika virus, human papillomavirus (HPV), and COVID-19, as well as safety, efficacy, and immunogenicity studies of DNA vaccines. Concurrently, the burst citation analysis identified emerging themes, including the development of DNA vaccines for COVID-19, Ebola, and MERS-CoV, as well as innovations in antigen design and delivery technologies. This study offers valuable insights into the evolution and future directions of DNA vaccine research, emphasizing its importance for global public health and the potential to address current and future health challenges.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Hematology, Jining NO. 1 People’ Hospital, Jining, China
| | - Haiguo Zhang
- Department of Hematology, Jining NO. 1 People’ Hospital, Jining, China
| | - Cuicui Yao
- Department of Hematology, Shandong Second Provincial General Hospital, Jinan, China
| | - Lihua Gu
- Department of Hematology, Jining NO. 1 People’ Hospital, Jining, China
| | - Shasha Dong
- Department of Hematology, Jining NO. 1 People’ Hospital, Jining, China
| | - Yamei Wu
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Lele Miao
- Department of Thyroid and Breast Surgery, Jining NO. 1 People’ Hospital, Jining, China
| |
Collapse
|
3
|
Konjengbam BD, Meitei HN, Pandey A, Haobam R. Goals and strategies in vaccine development against tuberculosis. Mol Immunol 2025; 183:56-71. [PMID: 40327952 DOI: 10.1016/j.molimm.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/16/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), continues to be a major health problem globally. The emergence of multi-drug-resistant TB and extensively drug-resistant TB has become a severe threat to TB control programs. Currently, the Bacille Calmette-Guerin (BCG) vaccine protects a child from disease dissemination efficiently, but its efficiency wanes in adults. Despite all the limitations of BCG and accelerated TB vaccine research, BCG remains the only approved vaccine available for TB. Anti-TB drug treatment has been successful in combating the disease, but it has various side effects and requires an extended drug treatment period. So, vaccination is the finest outlook that can surpass the above-mentioned limitations. Several vaccine candidates are in the pipeline, and the hope for a potential candidate to either boost the BCG vaccine or replace BCG is underway. This review discusses different approaches to TB vaccine development. It summarizes all the challenges and limitations in vaccine development, and its preclinical and clinical trials. Additionally, DNA vaccines and their vaccination techniques are also discussed. Furthermore, the immunoinformatics approach and nanomaterial-based vaccine delivery with practical and productive endpoints are also discussed. Lastly, the potential prospects are also suggested for further studies, which would help bring positive outcomes.
Collapse
Affiliation(s)
| | | | - Anupama Pandey
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur 795003, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur 795003, India.
| |
Collapse
|
4
|
Petrov GV, Koldina AM, Ledenev OV, Tumasov VN, Nazarov AA, Syroeshkin AV. Nanoparticles and Nanomaterials: A Review from the Standpoint of Pharmacy and Medicine. Pharmaceutics 2025; 17:655. [PMID: 40430945 PMCID: PMC12114779 DOI: 10.3390/pharmaceutics17050655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Nanoparticles (NPs) represent a unique class of structures in the modern world. In comparison to macro- and microparticles, NPs exhibit advantages due to their physicochemical properties. This has resulted in their extensive application not only in technical and engineering sciences, but also in pharmacy and medicine. A recent analysis of the scientific literature revealed that the number of articles related to the search term "nanoparticle drugs" has exceeded 65,000 in the last decade alone, according to PubMed. The growth of scientific publications on NPs and nanomaterials (NMs) in pharmacy demonstrates the rapidly developing interest of scientists in exploring alternative ways to deliver drugs, thereby improving their pharmacokinetic and pharmacodynamic properties, and the increased biocompatibility of many nanopharmaceuticals is a unique key to two mandatory pharmaceutical requirements-drug efficacy and safety. A comprehensive review of the literature indicates that the modern pharmaceutical industry is increasingly employing nanostructures. The exploration of their physicochemical properties with a subsequent modern approach to quality control remains the main task of modern pharmaceutical chemistry. The primary objective of this review is to provide a comprehensive overview of data on NPs, their physicochemical properties, and modern approaches to their synthesis, modification of their surface, and application in pharmacy.
Collapse
Affiliation(s)
- Gleb V. Petrov
- Department of Pharmaceutical and Toxicological Chemistry, Medical Institute, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.M.K.); (A.V.S.)
| | - Alena M. Koldina
- Department of Pharmaceutical and Toxicological Chemistry, Medical Institute, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.M.K.); (A.V.S.)
| | - Oleg V. Ledenev
- Department of Biology, Lomonosov Moscow State University, Leninskie Gory, 119234 Moscow, Russia;
| | - Vladimir N. Tumasov
- Department of Pharmaceutical Chemistry and Organization of Pharmaceutical Business, Faculty of Medicine, Lomonosov Moscow State University, GSP-1, Leninskie Gory, 119991 Moscow, Russia;
| | - Aleksandr A. Nazarov
- Department of Pharmaceutical and Toxicological Chemistry, Medical Institute, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.M.K.); (A.V.S.)
| | - Anton V. Syroeshkin
- Department of Pharmaceutical and Toxicological Chemistry, Medical Institute, RUDN University, 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.M.K.); (A.V.S.)
| |
Collapse
|
5
|
Martin C, Côté-Cyr M, Nguyen PT, Archambault D, Bourgault S. Evaluation of cylindrical micelles assembled from amphiphilic β-peptides as antigen delivery nanostructures. NANOSCALE ADVANCES 2025; 7:2979-2987. [PMID: 40177387 PMCID: PMC11960782 DOI: 10.1039/d5na00166h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
Supramolecular nanostructures assembled from synthetic peptides constitute promising scaffolds for the delivery of antigens for vaccine development. Amphiphilic peptides and self-assembling cross-β-peptides have been shown to promote cellular uptake of antigenic epitopes by antigen-presenting cells, to stimulate the innate immune system and to induce a robust antigen-specific humoral immune response. In this study, we evaluated the use of cylindrical micelles assembled from the amphiphilic β-peptide C16V3A3K3 as a vaccine nanoplatform, combining the properties of cross-β-sheet fibrils and micelles. The ectodomain of the matrix 2 protein (M2e) of the influenza A virus was conjugated with a tetra-Gly linker at the C-terminus of C16V3A3K3. The chimeric peptide assembled into biocompatible unbranched filaments that exposed the antigen on the surface, and these filaments were readily internalized by dendritic cells and activated the toll-like receptor 2/6. These cylindrical micelles induced a robust M2e-specific humoral immune response upon intramuscular immunization in mice without the need for co-administration with adjuvants. Although this strong humoral response did not translate into protection against a lethal infection with the H1N1 influenza virus, these cylindrical micelles assembled from amphiphilic β-peptides expand the repertoire of self-adjuvanted nanostructures to enhance antibody production against peptide epitopes.
Collapse
Affiliation(s)
- Clément Martin
- Department of Chemistry, Université du Québec à Montréal C.P.8888, Succursale Centre-Ville Montréal H3C 3P8 Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO) Québec H3C 3P8 Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA) Saint-Hyacinthe J2S 2M2 Canada
- Department of Biological Sciences, Université du Québec C.P.8888, Succursale Centre-Ville Montréal H3C 3P8 Canada
| | - Mélanie Côté-Cyr
- Department of Chemistry, Université du Québec à Montréal C.P.8888, Succursale Centre-Ville Montréal H3C 3P8 Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO) Québec H3C 3P8 Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA) Saint-Hyacinthe J2S 2M2 Canada
| | - Phuong Trang Nguyen
- Department of Chemistry, Université du Québec à Montréal C.P.8888, Succursale Centre-Ville Montréal H3C 3P8 Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO) Québec H3C 3P8 Canada
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA) Saint-Hyacinthe J2S 2M2 Canada
| | - Denis Archambault
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA) Saint-Hyacinthe J2S 2M2 Canada
- Department of Biological Sciences, Université du Québec C.P.8888, Succursale Centre-Ville Montréal H3C 3P8 Canada
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal C.P.8888, Succursale Centre-Ville Montréal H3C 3P8 Canada
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO) Québec H3C 3P8 Canada
- Department of Biological Sciences, Université du Québec C.P.8888, Succursale Centre-Ville Montréal H3C 3P8 Canada
| |
Collapse
|
6
|
Zaccariotto GDC, Bistaffa MJ, Zapata AMM, Rodero C, Coelho F, Quitiba JV, Lima L, Sterman R, Cardoso VDO, Zucolotto V. Cancer Nanovaccines: Mechanisms, Design Principles, and Clinical Translation. ACS NANO 2025; 19:16204-16223. [PMID: 40202241 PMCID: PMC12060653 DOI: 10.1021/acsnano.4c15765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
Cancer immunotherapy has transformed the landscape of oncological treatment by employing various strategies to teach the immune system to eliminate tumors. Among these, cancer nanovaccines are an emerging strategy that utilizes nanotechnology to enhance immune activation in response to tumor antigens. This review addresses the principles behind the different technologies in this field aimed at generating a robust and effective immune response. The diversity of strategies adopted for the design of nanovaccines is discussed, including the types of active agents, nanocarriers, their functionalizations, and the incorporation of adjuvants. Furthermore, strategies to optimize nanoparticle formulations to enhance the antigen presentation, target immune cells, and organs and promote strong and durable antitumor responses are explored. Finally, we analyze the current state of clinical application, highlighting ongoing clinical trials and the future potential of cancer nanovaccines. The insights presented in this review aim to guide future research and development efforts in the field, contributing to the advancement of more effective and targeted nanovaccines in the fight against cancer.
Collapse
Affiliation(s)
- Gabriel de Camargo Zaccariotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Maria Julia Bistaffa
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Angelica Maria Mazuera Zapata
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Camila Rodero
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Fernanda Coelho
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - João Victor
Brandão Quitiba
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Lorena Lima
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Raquel Sterman
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | | | - Valtencir Zucolotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| |
Collapse
|
7
|
Gomari MM, Ghantabpour T, Pourgholam N, Rostami N, Hatfield SM, Namazifar F, Abkhiz S, Eslami SS, Ramezanpour M, Darestanifarahani M, Astsaturov I, Bencherif SA. Breaking barriers: Smart vaccine platforms for cancer immunomodulation. Cancer Commun (Lond) 2025; 45:529-571. [PMID: 39901621 PMCID: PMC12067400 DOI: 10.1002/cac2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Despite significant advancements in cancer treatment, current therapies often fail to completely eradicate malignant cells. This shortfall underscores the urgent need to explore alternative approaches such as cancer vaccines. Leveraging the immune system's natural ability to target and kill cancer cells holds great therapeutic potential. However, the development of cancer vaccines is hindered by several challenges, including low stability, inadequate immune response activation, and the immunosuppressive tumor microenvironment, which limit their efficacy. Recent progress in various fields, such as click chemistry, nanotechnology, exosome engineering, and neoantigen design, offer innovative solutions to these challenges. These achievements have led to the emergence of smart vaccine platforms (SVPs), which integrate protective carriers for messenger ribonucleic acid (mRNA) with functionalization strategies to optimize targeted delivery. Click chemistry further enhances SVP performance by improving the encapsulation of mRNA antigens and facilitating their precise delivery to target cells. This review highlights the latest developments in SVP technologies for cancer therapy, exploring both their opportunities and challenges in advancing these transformative approaches.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | - Taha Ghantabpour
- Department of AnatomySchool of MedicineQazvin University of Medical SciencesQazvinIran
| | - Nima Pourgholam
- School of Nursing and MidwiferyIran University of Medical ScienceTehranIran
| | - Neda Rostami
- Department of Chemical EngineeringArak UniversityArakIran
| | - Stephen M. Hatfield
- New England Inflammation and Tissue Protection InstituteDepartment of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
- Department of Chemical EngineeringNortheastern UniversityBostonMassachusettsUSA
| | | | - Shadi Abkhiz
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | - Seyed Sadegh Eslami
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
- Molecular Proteomics LaboratoryBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - Mahsa Ramezanpour
- Department of Medical BiotechnologyFaculty of Allied MedicineIran University of Medical SciencesTehranIran
| | | | - Igor Astsaturov
- Marvin and Concetta Greenberg Pancreatic Cancer InstituteFox Chase Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Sidi A. Bencherif
- Department of Chemical EngineeringNortheastern UniversityBostonMassachusettsUSA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Polymers, Biopolymers Surfaces (PBS) LaboratoryNational Center for Scientific Research (CNRS) Mixed Research Unit (UMR) 6270University Rouen NormandieRouenFrance
| |
Collapse
|
8
|
Alemi PS, Mohamadali M, Arabahmadi S, Irani S, Sharifi F. Carboxymethyl Chitosan and Chitosan as a Bioactive Delivery System: A Review. Biotechnol Appl Biochem 2025:e2758. [PMID: 40275440 DOI: 10.1002/bab.2758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/16/2025] [Indexed: 04/26/2025]
Abstract
The functionality and mechanism of bioactive agents (BA) in treating various diseases have been studied as a progressive route. Designing an effective delivery system for transferring these molecules and components is a major challenge. For that reason, a wide range of biomaterials has been introduced to deliver BA to the target tissue or cells. Chitosan (CTS) is a nontoxic, biocompatible, biodegradable, and notable point low-cost polymer, and, as a result, can be effectively utilized in the formulation of diverse delivery systems, in biomedical applications. However, CTS has some limitations, such as poor solubility in aqueous and alkaline media, rapid swelling and degradation, and consequence fast release agent. The CTS derivative carboxymethyl chitosan (CMC) is an acceptable candidate for overcoming these limitations. CMC is a high-impact grade for pharmaceutical and biomedical applications because of its nontoxic, biocompatible, biodegradable, gelation, mucoadhesive, antibacterial, and antifungal. CMC bioactivity potentials are related to carboxyl and methyl groups added through chemical modification in the CTS backbone. In this review, the physical and chemical properties of CTS and CMC have been introduced and discussed. Afterward, its biomedical applications with delivery approaches for various BA (drugs, genes, proteins), microfluidic, and cancer have been considered.
Collapse
Affiliation(s)
- Parinaz Sadat Alemi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Marjan Mohamadali
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Samira Arabahmadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Fereshteh Sharifi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
- Soft Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
9
|
Sharma DK. Current advancements in nanoparticles for vaccines and drug delivery for the treatment of tuberculosis. J Microbiol Methods 2025; 232-234:107138. [PMID: 40280241 DOI: 10.1016/j.mimet.2025.107138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/05/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Since the early centuries until the present, tuberculosis has been a global illness with no cure. However, the sickness remains dormant in the afflicted individuals in certain circumstances. Due to the thick lipid mycobacterial wall and the challenging medication delivery into the bacterial cell, treatment is complex at this point. Over the past utics., there has been a growth in the function of nanomaterials in tuberculosis (TB) management, especially in the domains of early diagnosis, vaccine, and therapy. It has been demonstrated that nanomaterials are effective in quickly and accurately identifying tuberculosis germs. Additionally, novel nanocarriers have shown great promise for improved medication delivery and immunization, possibly increasing drug concentrations in target organs while lowering the frequency of treatments. Furthermore, the engineering of antigen carriers is a promising area of tuberculosis research that may lead to the successful creation of a novel class of potent TB vaccines. This article addresses tuberculosis infection diagnosis, pathophysiology, immunology, and advanced nanoparticles to deliver TB vaccines and anti-TB drugs. The challenges and prospects for the future in creating secure and functional nanoparticles for tuberculosis treatment and diagnosis for the good health and well-being of the patient are also discussed.
Collapse
Affiliation(s)
- Dinesh Kumar Sharma
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India.
| |
Collapse
|
10
|
Oubohssaine M, Hnini M, Rabeh K. Phospholipid signaling in plant growth and development: Insights, biotechnological implications and future directions. JOURNAL OF PLANT PHYSIOLOGY 2025; 307:154454. [PMID: 40015232 DOI: 10.1016/j.jplph.2025.154454] [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: 12/26/2024] [Revised: 02/03/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
Phospholipid signaling is essential for plant growth and development, orchestrating cellular membrane dynamics and regulating physiological processes critical for environmental adaptation. Phosphatidic acid (PA) plays diverse roles in key plant functions, including facilitating pollen tube growth, protecting against H2O2-induced cell death, and modulating actin cytoskeleton polymerization. Additionally, PA influences abscisic acid (ABA) signaling, impacting ionic flux, stomatal movement, and superoxide production. Phospholipase D (PLD) emerges as a crucial regulator, potentially linking and orchestrating microtubule reorganization. Saturated fatty acids, produced through phospholipase A (PLA) activity, also regulate various cellular processes. In Arabidopsis thaliana, Defender Against Apoptotic Death1 (DAD1), a plastidic PC-PLA1, supports jasmonic acid (JA) biosynthesis, which is essential for pollen maturation and flower development. Phospholipid signaling significantly influences stomatal function, with phospholipases modulating stomatal closure. This signaling pathway also plays a critical role in root development, where phosphocholine (PCho) and PA regulate root growth and tip growth of root hairs. This review highlights the pivotal role of phospholipid signaling pathways in coordinating plant growth, development, and responses to environmental cues. It explores the roles of PLD and PA in signal transduction and membrane degradation, particularly in seed aging. Additionally, it discusses the biotechnological applications of plant lipids, including genetic engineering for nutritional enhancement and biofuel production. Despite recent advancements, challenges such as low yield remain obstacles to the widespread adoption of biodiesel technology.
Collapse
Affiliation(s)
- Malika Oubohssaine
- Microbiology and Molecular Biology Team, Center of Plant and Microbial Biotechnology, Biodiversity and Environment, Faculty of Sciences, Mohammed V University in Rabat, Avenue Ibn Battouta, BP 1014, Rabat, 10000, Morocco.
| | - Mohamed Hnini
- Research Team in Science and Technology, High School of Technology Laayoune, Ibn Zohr University, Morocco
| | - Karim Rabeh
- Oasis System Research Unit, Regional Center of Agricultural Research of Errachidia, National Institute of Agricultural Research, PO. Box 415, Rabat, 10090, Morocco
| |
Collapse
|
11
|
Skwarczynski M, Alharbi N, Nahar UJ, Shalash AO, Azuar A, Koirala P, Khisty SJ, Wang J, Marasini N, Hussein WM, Khalil ZG, Toth I. Influence of component structural arrangement on cholesterol-antigen conjugate immunogenicity and antisera bactericidal activity against group A Streptococcus. Bioorg Chem 2025; 157:108248. [PMID: 39952060 DOI: 10.1016/j.bioorg.2025.108248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
Immune stimulants (adjuvants) are essential vaccine components; however, clinically approved adjuvants are limited with the majority being derived from pathogenic components. In this study, the adjuvanting capacity of cholesterol, a natural human lipid, was explored following conjugation with peptide antigens. A structure-activity relationship study was conducted to compare linear and branched cholesterol conjugates with other lipopeptide vaccines and commercial adjuvants. Group A Streptococcus (GAS) M protein-derived J8 B-cell epitope and a universal helper T-cell epitope P25 were selected as an antigen. In addition, liposomal formulations of the cholesterol-based vaccines were also evaluated in the mouse model. Following subcutaneous and intranasal administration, conjugates comprised of cholesterol, P25 and J8 induced the highest antibody production. Linear cholesterol peptide vaccines triggered strong antibody responses that killed GAS clinical isolates as effectively as responses triggered by commercial adjuvants. The immunogenicity of the vaccines was greatly influenced by the structural arrangement of the vaccine conjugate components. The lead cholesterol conjugate was self-adjuvanting and induced the desired immune response without any exogenous immune stimulation.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Nedaa Alharbi
- Applied College at Khulais, University of Jeddah, Jeddah 23890, Saudi Arabia
| | - Ummey J Nahar
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ahmed O Shalash
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Armira Azuar
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Prashamsa Koirala
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Shefali J Khisty
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jingwen Wang
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nirmal Marasini
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Waleed M Hussein
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zeinab G Khalil
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
12
|
Cui Z, Wang H, Qin L, Yuan Y, Xue J, An Y, Sun L, Zhu R, Li Q, Wang Y, Cui S, Zhan X, Zhai Q, Sun H, Zhang X, Guan J, Liu C, Mao S. Probing the Structural Elements of Polysaccharide Adjuvants for Enhancing Respiratory Mucosal Response: From Surmounting Multi-Obstacles to Eliciting Cascade Immunity. ACS NANO 2025; 19:11012-11028. [PMID: 40063734 DOI: 10.1021/acsnano.4c16788] [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: 03/26/2025]
Abstract
The immunomodulatory effects and excellent tolerability of polysaccharides make them optimal candidates for pulmonary vaccine adjuvants. Yet, the structure-immunostimulatory activity relationship of polysaccharides remains unrevealed. Here, we developed nanovaccines decorated with four polysaccharides of distinct structures─hyaluronic acid (HA), pectin (PC), chondroitin sulfate (SC), and heparan sulfate (SH)─all sharing similar particle sizes and zeta potential. Polysaccharides containing sulfate groups (SC, SH) exhibited superior efficacy in overcoming natural inhalation barriers and recruiting dendritic cells (DC). DC stimulation assays revealed that HA and SH significantly upregulated the expression of costimulation signals, with IL-6 secretion rising over 8.7-fold compared to pure OVA. Fluorescence resonance energy transfer demonstrated their detachment within the lysosomal microenvironment, thereby enhancing antigen cross-presentation. However, in vivo findings only showed that SH upregulated CCR7 chemokine and swiftly migrated to lymph nodes. Molecular docking and Western blot analyses further elucidated the involvement of the TLR─MyD88─TRAF6─NF-κB/MAPK/IRF-7 signaling pathways. Notably, SH-modified nanovaccines induced a more robust cellular and humoral immune response with the potential for immune memory. This study confirms that sulfate groups in polysaccharides enhance immune activation and that combining sulfate with acetyl groups offers a promising adjuvant configuration for augmenting mucosal, cellular, and humoral immunity.
Collapse
Affiliation(s)
- Zhixiang Cui
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hezhi Wang
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Qin
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ye Yuan
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingwen Xue
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yalin An
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Le Sun
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Renfang Zhu
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qingyu Li
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yi Wang
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuman Cui
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xuanguang Zhan
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiyao Zhai
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haiyan Sun
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhang
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Jian Guan
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Chang Liu
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Key Laboratory of Intelligent Mucosal Drug Delivery Systems, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| |
Collapse
|
13
|
Fernandes RS, de Assis Burle-Caldas G, Sergio SAR, Bráz AF, da Silva Leite NP, Pereira M, de Oliveira Silva J, Hojo-Souza NS, de Oliveira B, Fernandes APSM, da Fonseca FG, Gazzinelli RT, Dos Santos Ferreira D, Teixeira SMR. The immunogenic potential of an optimized mRNA lipid nanoparticle formulation carrying sequences from virus and protozoan antigens. J Nanobiotechnology 2025; 23:221. [PMID: 40102899 PMCID: PMC11921523 DOI: 10.1186/s12951-025-03201-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Lipid nanoparticles (LNP) are a safe and effective messenger RNA (mRNA) delivery system for vaccine applications, as shown by the COVID-19 mRNA vaccines. One of the main challenges faced during the development of these vaccines is the production of new and versatile LNP formulations capable of efficient encapsulation and delivery to cells in vivo. This study aimed to develop a new mRNA vaccine formulation that could potentially be used against existing diseases as well as those caused by pathogens that emerge every year. RESULTS Using firefly luciferase (Luc) as a reporter mRNA, we evaluated the physical-chemical properties, stability, and biodistribution of an LNP-mRNA formulation produced using a novel lipid composition and a microfluidic organic-aqueous precipitation method. Using mRNAs encoding a dengue virus or a Leishmania infantum antigen, we evaluated the immunogenicity of LNP-mRNA formulations and compared them with the immunization with the corresponding recombinant protein or plasmid-encoded antigens. For all tested LNP-mRNAs, mRNA encapsulation efficiency was higher than 85%, their diameter was around 100 nm, and their polydispersity index was less than 0.3. Following an intramuscular injection of 10 µg of the LNP-Luc formulation in mice, we detected luciferase activity in the injection site, as well as in the liver and spleen, as early as 6 h post-administration. LNPs containing mRNA encoding virus and parasite antigens were highly immunogenic, as shown by levels of antigen-specific IgG antibody as well as IFN-γ production by splenocytes of immunized animals that were similar to the levels that resulted from immunization with the corresponding recombinant protein or plasmid DNA. CONCLUSIONS Altogether, these results indicate that these novel LNP-mRNA formulations are highly immunogenic and may be used as novel vaccine candidates for different infectious diseases.
Collapse
Affiliation(s)
- Renata S Fernandes
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Gabriela de Assis Burle-Caldas
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | | | - Ana Flávia Bráz
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Nathália Pereira da Silva Leite
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Milton Pereira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Juliana de Oliveira Silva
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Pharmaceuticals, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Natália Satchiko Hojo-Souza
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
| | - Bianca de Oliveira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Ana Paula S Moura Fernandes
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Flávio Guimarães da Fonseca
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ricardo Tostes Gazzinelli
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Diego Dos Santos Ferreira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Pharmaceuticals, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Santuza M Ribeiro Teixeira
- Centro de Tecnologia de Vacinas da, Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil.
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
14
|
Chung MC, Mendez‐Gomez HR, Soni D, McGinley R, Zacharia A, Ashbrook J, Stover B, Grippin AJ, Sayour EJ, Guan J. Multi-Step Assembly of an RNA-Liposome Nanoparticle Formulation Revealed by Real-Time, Single-Particle Quantitative Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414305. [PMID: 39887619 PMCID: PMC11948016 DOI: 10.1002/advs.202414305] [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: 12/15/2024] [Indexed: 02/01/2025]
Abstract
Self-assembly plays a critical role in nanoparticle-based applications. However, it remains challenging to monitor the self-assembly of multi-component nanomaterials at a single-particle level, in real-time, with high throughput, and in a model-independent manner. Here, multi-color fluorescence microscopy is applied to track the assembly of both liposomes and mRNA simultaneously in clinical mRNA-based cancer immunotherapy. Imaging reveals that the assembly occurs in discrete steps: initially, RNA adsorbs onto the liposomes; then, the RNA-coated liposomes cluster into heterogeneous structures ranging from sub-micrometer to tens of micrometers. The clustering process is consistent with a Smoluchowski model with a Brownian diffusion kernel. The transition between the two steps of assembly is determined by the orientation of RNA-mediated interactions. Given the facile application of this approach and the ubiquity of the components studied, the imaging and analysis in this work are readily applied to monitor multi-component assembly of diverse nanomaterials.
Collapse
Affiliation(s)
- Michael C. Chung
- Division of Chemical Biology and Medicinal ChemistryCollege of PharmacyUniversity of Texas at AustinAustinTX78712USA
- Department of PhysicsUniversity of FloridaGainesvilleFL32611USA
| | - Hector R. Mendez‐Gomez
- Department of NeurosurgeryPreston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida Lillian S. WellsGainesvilleFL32610USA
| | - Dhruvkumar Soni
- Department of NeurosurgeryPreston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida Lillian S. WellsGainesvilleFL32610USA
| | - Reagan McGinley
- Department of Microbiology and Cell ScienceUniversity of FloridaGainesvilleFL32603USA
| | - Alen Zacharia
- Department of PhysicsUniversity of FloridaGainesvilleFL32611USA
| | - Jewel Ashbrook
- Middlebury College Department of PhysicsMcCardell Bicentennial HallMiddleburyVT05753USA
| | - Brian Stover
- Department of PediatricsDivision of Pediatric Hematology OncologyUniversity of FloridaGainesvilleFL32610USA
| | - Adam J. Grippin
- MD Anderson Cancer CenterDivision of Radiation OncologyUniversity of TexasHoustonTX77030USA
| | - Elias J. Sayour
- Department of NeurosurgeryPreston A. Wells, Jr. Center for Brain Tumor Therapy, McKnight Brain Institute, University of Florida Lillian S. WellsGainesvilleFL32610USA
- Department of PediatricsDivision of Pediatric Hematology OncologyUniversity of FloridaGainesvilleFL32610USA
| | - Juan Guan
- Division of Chemical Biology and Medicinal ChemistryCollege of PharmacyUniversity of Texas at AustinAustinTX78712USA
| |
Collapse
|
15
|
Imani S, Lv S, Qian H, Cui Y, Li X, Babaeizad A, Wang Q. Current innovations in mRNA vaccines for targeting multidrug-resistant ESKAPE pathogens. Biotechnol Adv 2025; 79:108492. [PMID: 39637949 DOI: 10.1016/j.biotechadv.2024.108492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/30/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
The prevalence of multidrug-resistant (MDR) ESKAPE pathogens, including Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa, represents a critical global public health challenge. In response, mRNA vaccines offer an adaptable and scalable platform for immunotherapy against ESKAPE pathogens by encoding specific antigens that stimulate B-cell-driven antibody production and CD8+ T-cell-mediated cytotoxicity, effectively neutralizing these pathogens and combating resistance. This review examines recent advancements and ongoing challenges in the development of mRNA vaccines targeting MDR ESKAPE pathogens. We explore antigen selection, the nuances of mRNA vaccine technology, and the complex interactions between bacterial infections and antibiotic resistance. By assessing the potential efficacy of mRNA vaccines and addressing key barriers to their paraclinical implementation, this review highlights the promising function of mRNA-based immunization in combating MDR ESKAPE pathogens.
Collapse
Affiliation(s)
- Saber Imani
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Shuojie Lv
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Hongbo Qian
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Yulan Cui
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - XiaoYan Li
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China
| | - Ali Babaeizad
- Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Qingjing Wang
- Key Laboratory of Artificial Organs and Computational Medicine of Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang, China.
| |
Collapse
|
16
|
Meng T, Gao T, Qiao F, Xu H, Yu N, Zuo W, Yang J. A VZV-gE subunit vaccine decorated with mPLA elicits protective cellular immmune responses against varicella-zoster virus. Int Immunopharmacol 2025; 147:114033. [PMID: 39799738 DOI: 10.1016/j.intimp.2025.114033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/03/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
Herpes zoster is an acute infectious skin disease caused by the reactivation of latent varicella-zoster virus, vaccination, such as subunit vaccine with good safety, can effectively prevent shingles through increasing immunity of the body. However, protein antigens are prone to degradation and inactivation, which alone is generally not sufficient to induce potent immune effect. In this study, the liposomal vaccine platform modified with mPLA (TLR4 agonist) was developed to improve the immunogenicity of glycoprotein E (VZV-gE) derived from herpes zoster virus. The thin-film dispersion and freeze-drying methods were employed to encapsulate VZV-gE against degradation, enhance liposomal stability, and achieve better redissolution effects with an optimized cryoprotectant. The in vitro results presented that mPLA could effectively enhance the uptake of VZV-gE with DC2.4. In vivo immune effect evaluation showed that the prepared subunit vaccines could induce stronger IgG, IgG1, and IgG2a antibody levels in the mouse serum, improving humoral immune effects. And the secretion levels of Th1 cytokines (IFN-γ, IL-2) and Th2 cytokines (IL-4, IL-10) in the splenocytes were significantly increased, inducing protective cellular immune responses. Overall, this work presented that combining immunomodulatory adjuvants decorated nanocarriers to develop subunit vaccine platforms was a promising strategy to prevent the occurrence of herpes zoster effectively.
Collapse
Affiliation(s)
- Tingting Meng
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China; Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ting Gao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Fangxia Qiao
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hongxia Xu
- Science and Technology Centers, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Na Yu
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Wenbao Zuo
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Jianhong Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
17
|
Clarissa EM, Karmacharya M, Choi H, Kumar S, Cho YK. Nature Inspired Delivery Vehicles for CRISPR-Based Genome Editing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2409353. [PMID: 39901476 DOI: 10.1002/smll.202409353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/16/2025] [Indexed: 02/05/2025]
Abstract
The advent of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-based genome editing technologies has opened up groundbreaking possibilities for treating a wide spectrum of genetic disorders and diseases. However, the success of these technologies relies heavily on the development of efficient and safe delivery systems. Among the most promising approaches are natural and synthetic nanocarrier-mediated delivery systems, including viral vectors, extracellular vesicles (EVs), engineered cellular membrane particles, liposomes, and various nanoparticles. These carriers enhance the efficacy of the CRISPR system by providing a unique combination of efficiency, specificity, and reduced immunogenicity. Synthetic carriers such as liposomes and nanoparticles facilitate CRISPR delivery with high reproducibility and customizable functions. Viral vectors, renowned for their high transduction efficiency and broad tropism, serve as powerful vehicles for delivering CRISPR components to various cell types. EVs, as natural carriers of RNA and proteins, offer a stealth mechanism to evade immune detection, allowing for the targeted delivery of genome editors with minimal off-target effects. Engineered cellular membrane particles further improve delivery by simulating the cellular environment, enhancing uptake, and minimizing immune response. This review explores the innovative integration of CRISPR genome editors with various nanocarrier systems, focusing on recent advancements, applications, and future directions in therapeutic genome editing.
Collapse
Affiliation(s)
- Elizabeth Maria Clarissa
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Mamata Karmacharya
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Hyunmin Choi
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Sumit Kumar
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Yoon-Kyoung Cho
- Center for Algorithmic and Robotized Synthesis, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| |
Collapse
|
18
|
da Silva EF, dos Santos FAL, Pires HM, Bastos LM, Ribeiro LNDM. Lipid Nanoparticles Carrying Essential Oils for Multiple Applications as Antimicrobials. Pharmaceutics 2025; 17:178. [PMID: 40006545 PMCID: PMC11859743 DOI: 10.3390/pharmaceutics17020178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Lipid nanoparticles (LNPs) are versatile delivery systems with high interest because they allow the release of hydrophobic and hydrophilic molecules, such as essential oils (EOs) and plant extracts. This review covers published works between 2019 and 2024 that have reported the use of essential EO-based LNPs with antimicrobial properties and applications in human and animal health, as well as biopesticides. In the human healthcare field, reports have addressed the effect of encapsulating EOs in lipid nanosystems with antiviral, antibacterial, antiprotozoal and antifungal activities. In animal care, this still needs to be more deeply explored while looking for more sustainable alternatives against different types of parasites that affect animal health. Overall, the antibacterial activities of LNPs carrying EOs are described as alternatives to the use of synthetic antibiotics. In the field of agriculture, studies showed that these approaches in the control of phytopathogens and other pests that affect food production. There is a growing demand for innovative and more sustainable technologies. However, there are still some challenges to be overcome in order to allow these innovations to reach the market.
Collapse
Affiliation(s)
| | | | | | - Luciana Machado Bastos
- Institute of Biotechnology, Federal University of Uberlandia, Uberlandia 38405-302, Brazil;
| | | |
Collapse
|
19
|
Nasr SS, Paul P, Loretz B, Lehr CM. Realizing time-staggered expression of nucleic acid-encoded proteins by co-delivery of messenger RNA and plasmid DNA on a single nanocarrier. Drug Deliv Transl Res 2024; 14:3339-3353. [PMID: 39009932 DOI: 10.1007/s13346-024-01668-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
Co-delivery of different protein-encoding polynucleotide species with varying expression kinetics of their therapeutic product will become a prominent requirement in the realm of combined nucleic acid(NA)-based therapies in the upcoming years. The current study explores the capacity for time-staggered expression of encoded proteins by simultaneous delivery of plasmid DNA (pDNA) in the core and mRNA on the shell of the same nanocarrier. The core is based on a Gelatin Type A-pDNA coacervate, thermally stabilized to form an irreversible nanogel stable enough for the deposition of cationic coats namely, protamine sulfate or LNP-related lipid mixtures. Only the protamine-coated nanocarriers remained colloidally stable following mRNA loading and could successfully co-transfect murine dendritic cell line DC2.4 with fluorescent reporter mRNA(mCherry) and pDNA (pAmCyan1). Further investigation of the protamine-coated nanosystem only, the transfection efficiency (percentage of transfected cells) and level of protein expression (mean fluorescence intensity, MFI) of mRNA and pDNA, simultaneously delivered by the same nanocarrier, were compared and kinetically assessed over 48 h in DC2.4 using flow cytometry. The onset of transfection for both nucleotides was initially delayed, with levels < 5% at 6 h. Thereafter, mRNA transfection reached 90% after 24 h and continued to slightly increase until 48 h. In contrast, pDNA transfection was clearly slower, reaching approximately 40% after 24 h, but continuing to increase to reach 94% at 48 h. The time course of protein expression (represented by MFI) for both NAs essentially followed that of transfection. Model-independent as well as model-dependent kinetic parameters applied to the data further confirmed such time-staggered expression of the two NA's where mRNA's rate of transfection and protein expression initially exceeded those of pDNA in the first 24 h of the experiment whereas the opposite was true during the second 24 h of the experiment where pDNA displayed the higher response rates. We expect that innovative nanocarriers capable of time-staggered co-delivery of different nucleotides could open new perspectives for multi-dosing, pulsatile or sustained expression of nucleic acid-based therapeutics in protein replacement, vaccination, and CRISPR-mediated gene editing scenarios.
Collapse
Affiliation(s)
- Sarah S Nasr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany.
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.
- Fischell Department of Bioengineering, University of Maryland, College Park, USA.
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Pascal Paul
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany.
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
20
|
Wang J, Zhao Z, Wang Q, Shi J, Wong DWC, Cheung JCW. Advancements in Nanoparticle-Based Adjuvants for Enhanced Tuberculosis Vaccination: A Review. Vaccines (Basel) 2024; 12:1335. [PMID: 39771997 PMCID: PMC11680411 DOI: 10.3390/vaccines12121335] [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: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective vaccines. Nanoparticle-based adjuvants represent a promising approach to enhancing tuberculosis vaccine efficacy. This review focuses on the advantages of nanoparticulate-loaded vaccines, emphasizing their ability to improve antigen delivery, safety, and immunogenicity. We discuss the various types of nanoparticles and their unique physicochemical properties that contribute to improved antigen delivery and sustained immune activation. Additionally, we highlight the advantages of nanoparticle-based adjuvants in inducing strong cellular and humoral immunity, enhancing vaccine stability, and reducing adverse effects. Finally, we address current challenges and future perspectives in the application of these novel adjuvants, emphasizing their potential to transform TB vaccine strategies and ultimately contribute to better global health outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Department of Clinical Laboratory, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430073, China
| | - Zian Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Quan Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Jingyu Shi
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Duo Wai-Chi Wong
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - James Chung-Wai Cheung
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
21
|
Archambault MJ, Tshibwabwa LM, Côté-Cyr M, Moffet S, Shiao TC, Bourgault S. Nanoparticles as Delivery Systems for Antigenic Saccharides: From Conjugation Chemistry to Vaccine Design. Vaccines (Basel) 2024; 12:1290. [PMID: 39591192 PMCID: PMC11598982 DOI: 10.3390/vaccines12111290] [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: 09/11/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Glycoconjugate vaccines have been effective in preventing numerous bacterial infectious diseases and have shown recent potential to treat cancers through active immunotherapy. Soluble polysaccharides elicit short-lasting immune responses and are usually covalently linked to immunogenic carrier proteins to enhance the antigen-specific immune response by stimulating T-cell-dependent mechanisms. Nonetheless, the conjugation of purified polysaccharides to carrier proteins complexifies vaccine production, and immunization with protein glycoconjugates can lead to the undesirable immunogenic interference of the carrier. Recently, the use of nanoparticles and nanoassemblies for the delivery of antigenic saccharides has gathered attention from the scientific community. Nanoparticles can be easily functionalized with a diversity of functionalities, including T-cell epitope, immunomodulator and synthetic saccharides, allowing for the modulation and polarization of the glycoantigen-specific immune response. Notably, the conjugation of glycan to nanoparticles protects the antigens from degradation and enhances their uptake by immune cells. Different types of nanoparticles, such as liposomes assembled from lipids, inorganic nanoparticles, virus-like particles and dendrimers, have been explored for glycovaccine design. The versatility of nanoparticles and their ability to induce robust immune responses make them attractive delivery platforms for antigenic saccharides. The present review aims at summarizing recent advancements in the use of nano-scaled systems for the delivery of synthetic glycoantigens. After briefly presenting the immunological mechanisms required to promote a robust immune response against antigenic saccharides, this review will offer an overview of the current trends in the nanoparticle-based delivery of glycoantigens.
Collapse
Affiliation(s)
- Marie-Jeanne Archambault
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montreal, QC H3C 3P8, Canada (L.M.T.)
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC H3C 3P8, Canada
- The Center of Excellence in Research on Orphan Diseases-Fondation Courtois (CERMO-FC), Montreal, QC H3C 3P8, Canada
| | - Laetitia Mwadi Tshibwabwa
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montreal, QC H3C 3P8, Canada (L.M.T.)
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC H3C 3P8, Canada
- The Center of Excellence in Research on Orphan Diseases-Fondation Courtois (CERMO-FC), Montreal, QC H3C 3P8, Canada
| | - Mélanie Côté-Cyr
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montreal, QC H3C 3P8, Canada (L.M.T.)
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC H3C 3P8, Canada
- The Center of Excellence in Research on Orphan Diseases-Fondation Courtois (CERMO-FC), Montreal, QC H3C 3P8, Canada
| | - Serge Moffet
- Glycovax Pharma Inc., Laval, QC H7V 5B7, Canada; (S.M.); (T.C.S.)
| | - Tze Chieh Shiao
- Glycovax Pharma Inc., Laval, QC H7V 5B7, Canada; (S.M.); (T.C.S.)
| | - Steve Bourgault
- Department of Chemistry, Université du Québec à Montréal, C.P.8888, Succursale Centre-Ville, Montreal, QC H3C 3P8, Canada (L.M.T.)
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Montreal, QC H3C 3P8, Canada
- The Center of Excellence in Research on Orphan Diseases-Fondation Courtois (CERMO-FC), Montreal, QC H3C 3P8, Canada
| |
Collapse
|
22
|
Chi WY, Hu Y, Huang HC, Kuo HH, Lin SH, Kuo CTJ, Tao J, Fan D, Huang YM, Wu AA, Hung CF, Wu TC. Molecular targets and strategies in the development of nucleic acid cancer vaccines: from shared to personalized antigens. J Biomed Sci 2024; 31:94. [PMID: 39379923 PMCID: PMC11463125 DOI: 10.1186/s12929-024-01082-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 10/10/2024] Open
Abstract
Recent breakthroughs in cancer immunotherapies have emphasized the importance of harnessing the immune system for treating cancer. Vaccines, which have traditionally been used to promote protective immunity against pathogens, are now being explored as a method to target cancer neoantigens. Over the past few years, extensive preclinical research and more than a hundred clinical trials have been dedicated to investigating various approaches to neoantigen discovery and vaccine formulations, encouraging development of personalized medicine. Nucleic acids (DNA and mRNA) have become particularly promising platform for the development of these cancer immunotherapies. This shift towards nucleic acid-based personalized vaccines has been facilitated by advancements in molecular techniques for identifying neoantigens, antigen prediction methodologies, and the development of new vaccine platforms. Generating these personalized vaccines involves a comprehensive pipeline that includes sequencing of patient tumor samples, data analysis for antigen prediction, and tailored vaccine manufacturing. In this review, we will discuss the various shared and personalized antigens used for cancer vaccine development and introduce strategies for identifying neoantigens through the characterization of gene mutation, transcription, translation and post translational modifications associated with oncogenesis. In addition, we will focus on the most up-to-date nucleic acid vaccine platforms, discuss the limitations of cancer vaccines as well as provide potential solutions, and raise key clinical and technical considerations in vaccine development.
Collapse
Affiliation(s)
- Wei-Yu Chi
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Yingying Hu
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hsin-Che Huang
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hui-Hsuan Kuo
- Pharmacology PhD Program, Weill Cornell Medicine, New York, NY, USA
| | - Shu-Hong Lin
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX, USA
| | - Chun-Tien Jimmy Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Julia Tao
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Darrell Fan
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Yi-Min Huang
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Annie A Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - T-C Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
Nahar UJ, Wang J, Shalash AO, Lu L, Islam MT, Alharbi N, Koirala P, Khalil ZG, Capon RJ, Hussein WM, Toth I, Skwarczynski M. Self-assembled monovalent lipidated mannose ligand as a standalone nanoadjuvant. Vaccine 2024; 42:126060. [PMID: 38897890 DOI: 10.1016/j.vaccine.2024.06.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/06/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024]
Abstract
Subunit vaccines require an immunostimulant (adjuvant) and/or delivery system to induce immunity. However, currently, available adjuvants are either too dangerous in terms of side effects for human use (experimental adjuvants) or have limited efficacy and applicability. In this study, we examined the capacity of mannose-lipopeptide ligands to enhance the immunogenicity of a vaccine consisting of polyleucine(L15)-antigen conjugates anchored to liposomes. The clinically tested Group A Streptococcus (GAS) B-cell epitope, J8, combined with universal T helper PADRE (P) was used as the antigen. Six distinct mannose ligands were incorporated into neutral liposomes carrying L15PJ8. While induced antibody titers were relatively low, the ligand carrying mannose, glycine/lysine spacer, and two palmitic acids as liposomal membrane anchoring moieties (ligand 3), induced significantly higher IgG titers than non-mannosylated liposomes. The IgG titers were significantly enhanced when positively charged liposomes were employed. Importantly, the produced antibodies were able to kill GAS bacteria. Unexpectedly, the physical mixture of only ligand 3 and PJ8 produced self-assembled nanorods that induced antibody titers as high as those elicited by the lead liposomal formulation and antigen adjuvanted with the potent, but toxic, complete Freund's adjuvant (CFA). Antibodies produced upon immunization with PJ8 + 3 were even more opsonic than those induced by CFA + PJ8. Importantly, in contrast to CFA, ligand 3 did not induce observable adverse reactions or excessive inflammatory responses. Thus, we demonstrated that a mannose ligand, alone, can serve as an effective vaccine nanoadjuvant.
Collapse
Affiliation(s)
- Ummey J Nahar
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jingwen Wang
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ahmed O Shalash
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Lantian Lu
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Md T Islam
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nedaa Alharbi
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Prashamsa Koirala
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zeinab G Khalil
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Robert J Capon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Waleed M Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia.
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
24
|
Kong W, Wei Y, Dong Z, Liu W, Zhao J, Huang Y, Yang J, Wu W, He H, Qi J. Role of size, surface charge, and PEGylated lipids of lipid nanoparticles (LNPs) on intramuscular delivery of mRNA. J Nanobiotechnology 2024; 22:553. [PMID: 39261807 PMCID: PMC11389890 DOI: 10.1186/s12951-024-02812-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 08/24/2024] [Indexed: 09/13/2024] Open
Abstract
Lipid nanoparticles (LNPs) are currently the most commonly used non-viral gene delivery system. Their physiochemical attributes, encompassing size, charge and surface modifications, significantly affect their behaviors both in vivo and in vitro. Nevertheless, the effects of these properties on the transfection and distribution of LNPs after intramuscular injection remain elusive. In this study, LNPs with varying sizes, lipid-based charges and PEGylated lipids were formulated to study their transfection and in vivo distribution. Luciferase mRNA (mLuc) was entraped in LNPs as a model nucleic acid molecule. Results indicated that smaller-sized LNPs and those with neutral potential presented superior transfection efficiency after intramuscular injection. Surprisingly, the sizes and charges did not exert a notable influence on the in vivo distribution of the LNPs. Furthermore, PEGylated lipids with shorter acyl chains contributed to enhanced transfection efficiency due to their superior cellular uptake and lysosomal escape capabilities. Notably, the mechanisms underlying cellular uptake differed among LNPs containing various types of PEGylated lipids, which was primarily attributed to the length of their acyl chain. Together, these insights underscore the pivotal role of nanoparticle characteristics and PEGylated lipids in the intramuscular route. This study not only fills crucial knowledge gaps but also provides significant directions for the effective delivery of mRNA via LNPs.
Collapse
Affiliation(s)
- Weiwen Kong
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China
| | - Yuning Wei
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China
| | - Zirong Dong
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China
| | - Wenjuan Liu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China
| | - Jiaxin Zhao
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China
| | - Yan Huang
- Department of Oncology, Shanghai Medical College of Fudan University, 270 Dong-an Road, Shanghai, 200032, China
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China
| | - Jinlong Yang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China
| | - Wei Wu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China
| | - Haisheng He
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China.
| | - Jianping Qi
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai, 201203, China.
| |
Collapse
|
25
|
Burgos JM, Vega E, García ML, Pujol M, Sánchez-López E, Souto EB. Biodegradable nanoplatforms for antigen delivery: part II - nanoparticles, hydrogels, and microneedles for cancer immunotherapy. Expert Opin Drug Deliv 2024; 21:1385-1394. [PMID: 39245925 DOI: 10.1080/17425247.2024.2400291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION In recent years, chimeric antigen receptor T (CAR-T) cell therapy has resulted in a breakthrough in the treatment of patients with refractory or relapsed hematological malignancies. However, the identification of patients suitable for CAR-T cell therapy needs to be improved. AREASCOVERED CAR-T cell therapy has demonstrated excellent efficacy in hematological malignancies; however, views on determining when to apply CAR-T cells in terms of the evaluation of patient characteristics remain controversial. EXPERT OPINION We reviewed the current feasibility and challenges of CAR-T cell therapy in the most common hematological malignancies and classified them according to disease type and treatment priority, to guide clinicians and researchers in applying and investigating CAR-T cells further.
Collapse
Affiliation(s)
- Jordi Madariaga Burgos
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Estefanía Vega
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - María Luisa García
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Montserrat Pujol
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
26
|
Kane GI, Brassil ML, Diaz-Infante MB, Atukorale PU. Nanocarrier design for pathogen-inspired innate immune agonist delivery. Trends Immunol 2024; 45:678-692. [PMID: 39191543 PMCID: PMC11492413 DOI: 10.1016/j.it.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024]
Abstract
In complex diseases such as cancer, modulating cytokine signatures of disease using innate immune agonists holds therapeutic promise. Novel multi-agonist treatments offer tunable control of the immune system because they are uniquely pathogen inspired, eliciting robust antitumor responses by promoting synergistic cytokine responses. However, the chief strategic hurdle is ensuring multi-agonist delivery to the same target cells, highlighting the importance of using nanomaterial-based carriers. Here, we place nanocarriers in center stage and review the delivery hurdles related to the varying extra- and intracellular localizations of innate immune receptors. We discuss a range of nanomaterials used for multi-agonist delivery, highlighting their respective benefits and drawbacks. Our overarching stance is that rational nanocarrier design is crucial for developing pathogen-inspired multi-agonist immunotherapies.
Collapse
Affiliation(s)
- Griffin I Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA; UMass Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Meghan L Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA; UMass Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Miranda B Diaz-Infante
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA; UMass Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Prabhani U Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA; Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; UMass Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
27
|
Shamseldin MM, Read KA, Hall JM, Tuazon JA, Brown JM, Guo M, Gupta YA, Deora R, Oestreich KJ, Dubey P. The adjuvant BcfA activates antigen presenting cells through TLR4 and supports T FH and T H1 while attenuating T H2 gene programming. Front Immunol 2024; 15:1439418. [PMID: 39267766 PMCID: PMC11390363 DOI: 10.3389/fimmu.2024.1439418] [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: 05/27/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Introduction Adjuvants added to subunit vaccines augment antigen-specific immune responses. One mechanism of adjuvant action is activation of pattern recognition receptors (PRRs) on innate immune cells. Bordetella colonization factor A (BcfA); an outer membrane protein with adjuvant function, activates TH1/TH17-polarized immune responses to protein antigens from Bordetella pertussis and SARS CoV-2. Unlike other adjuvants, BcfA does not elicit a TH2 response. Methods To understand the mechanism of BcfA-driven TH1/TH17 vs. TH2 activation, we screened PRRs to identify pathways activated by BcfA. We then tested the role of this receptor in the BcfA-mediated activation of bone marrow-derived dendritic cells (BMDCs) using mice with germline deletion of TLR4 to quantify upregulation of costimulatory molecule expression and cytokine production in vitro and in vivo. Activity was also tested on human PBMCs. Results PRR screening showed that BcfA activates antigen presenting cells through murine TLR4. BcfA-treated WT BMDCs upregulated expression of the costimulatory molecules CD40, CD80, and CD86 and produced IL-6, IL-12/23 p40, and TNF-α while TLR4 KO BMDCs were not activated. Furthermore, human PBMCs stimulated with BcfA produced IL-6. BcfA-stimulated murine BMDCs also exhibited increased uptake of the antigen DQ-OVA, supporting a role for BcfA in improving antigen presentation to T cells. BcfA further activated APCs in murine lungs. Using an in vitro TH cell polarization system, we found that BcfA-stimulated BMDC supernatant supported TFH and TH1 while suppressing TH2 gene programming. Conclusions Overall, these data provide mechanistic understanding of how this novel adjuvant activates immune responses.
Collapse
Affiliation(s)
- Mohamed M. Shamseldin
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Departments of Microbiology, The Ohio State University, Columbus, OH, United States
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University-Ain Helwan, Helwan, Egypt
| | - Kaitlin A. Read
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Jesse M. Hall
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Jasmine A. Tuazon
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Jessica M. Brown
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Myra Guo
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Yash A. Gupta
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Rajendar Deora
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Departments of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Kenneth J. Oestreich
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
| | - Purnima Dubey
- Departments of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
28
|
Parvin N, Joo SW, Mandal TK. Enhancing Vaccine Efficacy and Stability: A Review of the Utilization of Nanoparticles in mRNA Vaccines. Biomolecules 2024; 14:1036. [PMID: 39199422 PMCID: PMC11353004 DOI: 10.3390/biom14081036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/12/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
The development of vaccines has entered a new era with the advent of nanotechnology, particularly through the utilization of nanoparticles. This review focuses on the role of nanoparticles in enhancing the efficacy and stability of mRNA vaccines. Nanoparticles, owing to their unique properties such as high surface area, tunable size, and their ability to be functionalized, have emerged as powerful tools in vaccine development. Specifically, lipid nanoparticles (LNPs) have revolutionized the delivery of mRNA vaccines by protecting the fragile mRNA molecules and facilitating their efficient uptake by cells. This review discusses the various types of nanoparticles employed in mRNA vaccine formulations, including lipid-based, polymer-based, and inorganic nanoparticles, highlighting their advantages and limitations. Moreover, it explores the mechanisms by which nanoparticles improve immune responses, such as enhanced antigen presentation and the prolonged release of mRNA. This review also addresses the challenges and future directions in nanoparticle-based vaccine development, emphasizing the need for further research to optimize formulations for broader applications. By providing an in-depth analysis of the current advancements in and potential of nanoparticles in mRNA vaccines, this review aims to shed light on their critical role in combating infectious diseases and improving public health outcomes.
Collapse
Affiliation(s)
| | - Sang Woo Joo
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Tapas Kumar Mandal
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
29
|
Desai N, Chavda V, Singh TRR, Thorat ND, Vora LK. Cancer Nanovaccines: Nanomaterials and Clinical Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401631. [PMID: 38693099 DOI: 10.1002/smll.202401631] [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: 03/01/2024] [Revised: 03/30/2024] [Indexed: 05/03/2024]
Abstract
Cancer nanovaccines represent a promising frontier in cancer immunotherapy, utilizing nanotechnology to augment traditional vaccine efficacy. This review comprehensively examines the current state-of-the-art in cancer nanovaccine development, elucidating innovative strategies and technologies employed in their design. It explores both preclinical and clinical advancements, emphasizing key studies demonstrating their potential to elicit robust anti-tumor immune responses. The study encompasses various facets, including integrating biomaterial-based nanocarriers for antigen delivery, adjuvant selection, and the impact of nanoscale properties on vaccine performance. Detailed insights into the complex interplay between the tumor microenvironment and nanovaccine responses are provided, highlighting challenges and opportunities in optimizing therapeutic outcomes. Additionally, the study presents a thorough analysis of ongoing clinical trials, presenting a snapshot of the current clinical landscape. By curating the latest scientific findings and clinical developments, this study aims to serve as a comprehensive resource for researchers and clinicians engaged in advancing cancer immunotherapy. Integrating nanotechnology into vaccine design holds immense promise for revolutionizing cancer treatment paradigms, and this review provides a timely update on the evolving landscape of cancer nanovaccines.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502285, India
| | - Vivek Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, 380009, India
| | | | - Nanasaheb D Thorat
- Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
- Department of Physics, Bernal Institute, Castletroy, Limerick, V94T9PX, Ireland
- Nuffield Department of Women's & Reproductive Health, Medical Science Division, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| |
Collapse
|
30
|
Nanda S, Zafar MA, Lamba T, Malik JA, Khan MA, Bhardwaj P, Bisht B, Ghadi R, Kaur G, Bhalla V, Owais M, Jain S, Sehrawat S, Agrewala JN. A novel strategy to elicit enduring anti-morphine immunity and relief from addiction by targeting Acr1 protein nano vaccine through TLR-2 to dendritic cells. Int J Biol Macromol 2024; 274:133188. [PMID: 38880456 DOI: 10.1016/j.ijbiomac.2024.133188] [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: 05/22/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Morphine addiction poses a significant challenge to global healthcare. Current opioid substitution therapies, such as buprenorphine, naloxone and methadone are effective but often lead to dependence. Thus, exploring alternative treatments for opioid addiction is crucial. We have developed a novel vaccine that presents morphine and Pam3Cys (a TLR-2 agonist) on the surface of Acr1 nanoparticles. This vaccine has self-adjuvant properties and targets TLR-2 receptors on antigen-presenting cells, particularly dendritic cells. Our vaccination strategy promotes the proliferation and differentiation of morphine-specific B-cells and Acr1-reactive CD4 T-cells. Additionally, the vaccine elicited the production of high-affinity anti-morphine antibodies, effectively eliminating morphine from the bloodstream and brain in mice. It also reduced the expression of addiction-associated μ-opioid receptor and dopamine genes. The significant increase in memory CD4 T-cells and B-cells indicates the vaccine's ability to induce long-lasting immunity against morphine. This vaccine holds promise as a prophylactic measure against morphine addiction.
Collapse
Affiliation(s)
- Sidhanta Nanda
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Mohammad Adeel Zafar
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Taruna Lamba
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Jonaid Ahmad Malik
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Mohammad Affan Khan
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Priya Bhardwaj
- CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Bhawana Bisht
- CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Gurpreet Kaur
- Department of Biotechnology, Chandigarh Group of Colleges, Mohali, India
| | | | - Mohammad Owais
- Department of Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Javed N Agrewala
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India.
| |
Collapse
|
31
|
Maji M, Ghosh S, Didwania N, Ali N. Differentially Charged Liposomes Stimulate Dendritic Cells with Varying Effects on Uptake and Processing When Used Alone or in Combination with an Adjuvant. ACS OMEGA 2024; 9:29175-29185. [PMID: 39005780 PMCID: PMC11238303 DOI: 10.1021/acsomega.3c07814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/08/2024] [Accepted: 02/05/2024] [Indexed: 07/16/2024]
Abstract
Liposomes carrying differential charges have been extensively studied for their role in stimulating dendritic cells (DCs), major antigen-presenting cells, known to serve as a pivotal bridge between innate and adaptive immunity. However, the impact of the differentially charged liposomes on activating DCs remains to be understood. In this study, we have investigated the impact of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC)-based neutral, anionic, and cationic liposomes on the uptake, immunostimulation, and intracellular fate in mouse bone-marrow-derived DCs. We observed that liposomes could induce phenotypic maturation of DCs by inducing the expression of costimulatory molecules (CD40 and CD86) and production of cytokines tumor necrosis factor-α, interleukin-12,and nitric oxide. Interestingly, admixing monophosphoryl lipid A with charged liposomes further enhances the expression of the costimulatory molecules and production of cytokines, with preferential activation by positively charged liposomes. Fluorometric analysis using a pH-sensitive dye and flow-cytometry-based pathway inhibition assays revealed that cationic liposomes were taken up more efficiently by DCs through endocytosis and transported to neutral compartments for further processing, whereas anionic and neutral liposomes were inclined to accumulate in acidic compartments. These findings therefore endorse the use of cationic DSPC liposomes as a preferred option for vaccine delivery vehicles over neutral and negatively charged liposomes, particularly for the preferential activation of DCs.
Collapse
Affiliation(s)
| | | | - Nicky Didwania
- Infectious Diseases and Immunology
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Nahid Ali
- Infectious Diseases and Immunology
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
32
|
Shetty S, Alvarado PC, Pettie D, Collier JH. Next-Generation Vaccine Development with Nanomaterials: Recent Advances, Possibilities, and Challenges. Annu Rev Biomed Eng 2024; 26:273-306. [PMID: 38959389 DOI: 10.1146/annurev-bioeng-110122-124359] [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: 07/05/2024]
Abstract
Nanomaterials are becoming important tools for vaccine development owing to their tunable and adaptable nature. Unique properties of nanomaterials afford opportunities to modulate trafficking through various tissues, complement or augment adjuvant activities, and specify antigen valency and display. This versatility has enabled recent work designing nanomaterial vaccines for a broad range of diseases, including cancer, inflammatory diseases, and various infectious diseases. Recent successes of nanoparticle vaccines during the coronavirus disease 2019 (COVID-19) pandemic have fueled enthusiasm further. In this review, the most recent developments in nanovaccines for infectious disease, cancer, inflammatory diseases, allergic diseases, and nanoadjuvants are summarized. Additionally, challenges and opportunities for clinical translation of this unique class of materials are discussed.
Collapse
Affiliation(s)
- Shamitha Shetty
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Pablo Cordero Alvarado
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Deleah Pettie
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| |
Collapse
|
33
|
Jiang M, Zhang GH, Yu Y, Zhao YH, Liu J, Zeng Q, Feng MY, Ye F, Xiong DS, Wang L, Zhang YN, Yu L, Wei JJ, He LB, Zhi W, Du XR, Li NJ, Han CL, Yan HQ, Zhou ZT, Miao YB, Wang W, Liu WX. De novo design of a nanoregulator for the dynamic restoration of ovarian tissue in cryopreservation and transplantation. J Nanobiotechnology 2024; 22:330. [PMID: 38862987 PMCID: PMC11167790 DOI: 10.1186/s12951-024-02602-5] [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: 03/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
The cryopreservation and transplantation of ovarian tissue underscore its paramount importance in safeguarding reproductive capacity and ameliorating reproductive disorders. However, challenges persist in ovarian tissue cryopreservation and transplantation (OTC-T), including the risk of tissue damage and dysfunction. Consequently, there has been a compelling exploration into the realm of nanoregulators to refine and enhance these procedures. This review embarks on a meticulous examination of the intricate anatomical structure of the ovary and its microenvironment, thereby establishing a robust groundwork for the development of nanomodulators. It systematically categorizes nanoregulators and delves deeply into their functions and mechanisms, meticulously tailored for optimizing ovarian tissue cryopreservation and transplantation. Furthermore, the review imparts valuable insights into the practical applications and obstacles encountered in clinical settings associated with OTC-T. Moreover, the review advocates for the utilization of microbially derived nanomodulators as a potent therapeutic intervention in ovarian tissue cryopreservation. The progression of these approaches holds the promise of seamlessly integrating nanoregulators into OTC-T practices, thereby heralding a new era of expansive applications and auspicious prospects in this pivotal domain.
Collapse
Affiliation(s)
- Min Jiang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Guo-Hui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Yuan Yu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yu-Hong Zhao
- School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, China
| | - Jun Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Qin Zeng
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Meng-Yue Feng
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Fei Ye
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Dong-Sheng Xiong
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Li Wang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Ya-Nan Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Ling Yu
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Jia-Jing Wei
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Li-Bing He
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Weiwei Zhi
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Xin-Rong Du
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ning-Jing Li
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Chang-Li Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - He-Qiu Yan
- School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, China
| | - Zhuo-Ting Zhou
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Wen Wang
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Wei-Xin Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China.
| |
Collapse
|
34
|
Watanabe S, Tamura Y, Oba K, Kitayama S, Sato M, Kodera R, Toyoshima K, Chiba Y, Araki A. Hypopituitarism with secondary adrenocortical insufficiency and arginine vasopressin deficiency due to hypophysitis after COVID-19 vaccination: a case report. BMC Endocr Disord 2024; 24:71. [PMID: 38769570 PMCID: PMC11103972 DOI: 10.1186/s12902-024-01582-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Although vaccination against coronavirus disease (COVID-19) has several side effects, hypopituitarism due to hypophysitis has rarely been reported. CASE PRESENTATION An 83-year-old healthy woman, who had received her fourth COVID-19 vaccine dose 2 days before admission, presented to the emergency department with difficulty moving. On examination, impaired consciousness (Glasgow Coma Scale: 14) and fever were observed. Computed tomography and magnetic resonance imaging of the head revealed swelling from the sella turcica to the suprasellar region. Her morning serum cortisol level was low (4.4 μg/dL) and adrenocorticotropic hormone level was normal (21.6 pg/mL). Central hypothyroidism was also suspected (thyroid stimulating hormone, 0.46 μIU/mL; free triiodothyronine, 1.86 pg/mL; free thyroxine, 0.48 ng/dL). Secondary adrenocortical insufficiency, growth hormone deficiency, delayed gonadotropin response, and elevated prolactin levels were also observed. After administration of prednisolone and levothyroxine, her consciousness recovered. On the 7th day of admission, the patient developed polyuria, and arginine vasopressin deficiency was diagnosed using a hypertonic saline test. On the 15th day, the posterior pituitary gland showed a loss of high signal intensity and the polyuria resolved spontaneously. On the 134th day, the corticotropin-releasing hormone loading test showed a normal response; however, the thyrotropin-releasing hormone stimulation test showed a low response. The patient's disease course was stable with continued thyroid and adrenal corticosteroid supplementation. CONCLUSIONS Herein, we report a rare case of anterior hypopituitarism and arginine vasopressin deficiency secondary to hypophysitis following COVID-19 vaccination.
Collapse
Affiliation(s)
- So Watanabe
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Yoshiaki Tamura
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan.
| | - Kazuhito Oba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Saori Kitayama
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Motoya Sato
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Remi Kodera
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Kenji Toyoshima
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Yuko Chiba
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, 35-2 Sakaecho, Itabashi-Ku, Tokyo, 173-0015, Japan
| |
Collapse
|
35
|
Bonam SR, Hazell NC, Mathew MJ, Liang Y, Zhang X, Wei Z, Alameh MG, Weissman D, Hu H. Innate and Adaptive Immune Parameters following mRNA Vaccination in Mice. Vaccines (Basel) 2024; 12:543. [PMID: 38793794 PMCID: PMC11125668 DOI: 10.3390/vaccines12050543] [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: 04/13/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
The COVID-19 pandemic has raised the standard regarding the current vaccine development pace, as several messenger RNA (mRNA)-lipid nanoparticle (LNP) vaccines have proved their ability to induce strong immunogenicity and protective efficacy. We developed 1-methylpseudouridine-containing mRNA-LNP vaccines, expressing either the more conserved SARS-CoV-2 nucleoprotein (mRNA-N) or spike protein (mRNA-S), both based on the prototypic viral sequences. When combining both mRNA-S and mRNA-N together (mRNA-S+N), the vaccine showed high immunogenicity and broad protection against different SARS-CoV-2 variants, including wildtype, Delta, BA.1, BA.5, and BQ.1. To better understand the mechanisms behind this broad protection obtained by mRNA-S+N, we analyzed innate and adaptive immune parameters following vaccination in mice. Compared to either mRNA-S or mRNA-N alone, mice vaccinated with mRNA-S+N exhibited an increase in the innate immune response, as depicted by the higher cytokine (IL-6 and chemokine (MCP-1) levels. In addition, lymph node immunophenotyping showed the maturation and activation of dendritic cells and natural killer cells, respectively. To understand the adaptive immune response, RNA-Seq analyses of the lung and spleen samples of the vaccinated mice were performed in parallel and revealed a stronger immune gene-expression profile in the lung than that in the spleen. Compared to mRNA-S alone, mRNA-S+N vaccination elicited higher levels of expression for genes involved in multiple immune pathways, including T cells, cytokine signaling, antigen presentation, B cells, and innate immunity. Together, our studies provide immunological insights into the mechanisms of broad protection conferred by dual mRNA vaccination against SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (N.C.H.); (Y.L.)
| | - Nicholas C. Hazell
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (N.C.H.); (Y.L.)
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mano Joseph Mathew
- EFREI Research Lab, Panthéon Assas University, 30-32 Avenue de la République, 94800 Villejuif, France;
- Laboratoire Génomique, Bioinformatique et Chimie Moléculaire, EA7528, Conservatoire National des Arts et Métiers, HESAM Université, 2 rue Conté, 75003 Paris, France
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (N.C.H.); (Y.L.)
| | - Xuxiang Zhang
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA; (X.Z.); (Z.W.)
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA; (X.Z.); (Z.W.)
| | - Mohamad-Gabriel Alameh
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA; (M.-G.A.); (D.W.)
| | - Drew Weissman
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA; (M.-G.A.); (D.W.)
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (N.C.H.); (Y.L.)
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
36
|
Zhao M, He C, Zheng X, Jiang M, Xie Z, Wei H, Zhang S, Lin Y, Zhang J, Sun X. Self-adjuvanting polymeric nanovaccines enhance IFN production and cytotoxic T cell response. J Control Release 2024; 369:556-572. [PMID: 38580136 DOI: 10.1016/j.jconrel.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
Vaccines represent one of the most powerful and cost-effective innovations for controlling a wide range of infectious diseases caused by various viruses and bacteria. Unlike mRNA and DNA-based vaccines, subunit vaccines carry no risk of insertional mutagenesis and can be lyophilized for convenient transportation and long-term storage. However, existing adjuvants are often associated with toxic effect and reactogenicity, necessitating expanding the repertoire of adjuvants with better biocompatibility, for instance, designing self-adjuvating polymeric carriers. We herein report a novel subunit vaccine delivery platform constructed via in situ free radical polymerization of C7A (2-(Hexamethyleneimino) ethyl methacrylate) and acrylamide around the surface of individual protein antigens. Using ovalbumin (OVA) as a model antigen, we observed substantial increases in both diameter (∼70 nm) and surface potential (-1.18 mV) following encapsulation, referred to as n(OVA)C7A. C7A's ultra pH sensitivity with a transition pH around 6.9 allows for rapid protonation in acidic environments. This property facilitates crucial processes such as endosomal escape and major histocompatibility complex (MHC)-I-mediated antigen presentation, culminating in the substantial CD8+ T cell activation. Additionally, compared to OVA nanocapsules without the C7A components and native OVA without modifications, we observed heightened B cell activation within the germinal center, along with remarkable increases in serum antibody and cytokine production. It's important to note that mounting evidence suggests that adjuvant effects, particularly its targeted stimulation of type I interferons (IFNs), can contribute to advantageous adaptive immune responses. Beyond its exceptional potency, the nanovaccine also demonstrated robust formation of immune memory and exhibited a favorable biosafety profile. These findings collectively underscore the promising potential of our nanovaccine in the realm of immunotherapy and vaccine development.
Collapse
Affiliation(s)
- Ming Zhao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chunting He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Xueyun Zheng
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Min Jiang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Zhiqiang Xie
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Hongjiao Wei
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Shujun Zhang
- School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, PR China
| | - Ying Lin
- Guangdong Key Laboratory of Fermentation and Enzyme Engineering, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China.
| | - Jiaheng Zhang
- School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, PR China.
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
37
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
38
|
Sun Z, Zhao H, Ma L, Shi Y, Ji M, Sun X, Ma D, Zhou W, Huang T, Zhang D. The quest for nanoparticle-powered vaccines in cancer immunotherapy. J Nanobiotechnology 2024; 22:61. [PMID: 38355548 PMCID: PMC10865557 DOI: 10.1186/s12951-024-02311-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] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Despite recent advancements in cancer treatment, this disease still poses a serious threat to public health. Vaccines play an important role in preventing illness by preparing the body's adaptive and innate immune responses to combat diseases. As our understanding of malignancies and their connection to the immune system improves, there has been a growing interest in priming the immune system to fight malignancies more effectively and comprehensively. One promising approach involves utilizing nanoparticle systems for antigen delivery, which has been shown to potentiate immune responses as vaccines and/or adjuvants. In this review, we comprehensively summarized the immunological mechanisms of cancer vaccines while focusing specifically on the recent applications of various types of nanoparticles in the field of cancer immunotherapy. By exploring these recent breakthroughs, we hope to identify significant challenges and obstacles in making nanoparticle-based vaccines and adjuvants feasible for clinical application. This review serves to assess recent breakthroughs in nanoparticle-based cancer vaccinations and shed light on their prospects and potential barriers. By doing so, we aim to inspire future immunotherapies for cancer that harness the potential of nanotechnology to deliver more effective and targeted treatments.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hui Zhao
- Department of Endodontics, East Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Li Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yanli Shi
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Mei Ji
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Dan Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Wei Zhou
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Tao Huang
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Dongsheng Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
39
|
Shi Y, Zhen X, Zhang Y, Li Y, Koo S, Saiding Q, Kong N, Liu G, Chen W, Tao W. Chemically Modified Platforms for Better RNA Therapeutics. Chem Rev 2024; 124:929-1033. [PMID: 38284616 DOI: 10.1021/acs.chemrev.3c00611] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
RNA-based therapies have catalyzed a revolutionary transformation in the biomedical landscape, offering unprecedented potential in disease prevention and treatment. However, despite their remarkable achievements, these therapies encounter substantial challenges including low stability, susceptibility to degradation by nucleases, and a prominent negative charge, thereby hindering further development. Chemically modified platforms have emerged as a strategic innovation, focusing on precise alterations either on the RNA moieties or their associated delivery vectors. This comprehensive review delves into these platforms, underscoring their significance in augmenting the performance and translational prospects of RNA-based therapeutics. It encompasses an in-depth analysis of various chemically modified delivery platforms that have been instrumental in propelling RNA therapeutics toward clinical utility. Moreover, the review scrutinizes the rationale behind diverse chemical modification techniques aiming at optimizing the therapeutic efficacy of RNA molecules, thereby facilitating robust disease management. Recent empirical studies corroborating the efficacy enhancement of RNA therapeutics through chemical modifications are highlighted. Conclusively, we offer profound insights into the transformative impact of chemical modifications on RNA drugs and delineates prospective trajectories for their future development and clinical integration.
Collapse
Affiliation(s)
- Yesi Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xueyan Zhen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yiming Zhang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310058, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
40
|
Hemmati S, Saeidikia Z, Seradj H, Mohagheghzadeh A. Immunomodulatory Peptides as Vaccine Adjuvants and Antimicrobial Agents. Pharmaceuticals (Basel) 2024; 17:201. [PMID: 38399416 PMCID: PMC10892805 DOI: 10.3390/ph17020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
The underdevelopment of adjuvant discovery and diversity, compared to core vaccine technology, is evident. On the other hand, antibiotic resistance is on the list of the top ten threats to global health. Immunomodulatory peptides that target a pathogen and modulate the immune system simultaneously are promising for the development of preventive and therapeutic molecules. Since investigating innate immunity in insects has led to prominent achievements in human immunology, such as toll-like receptor (TLR) discovery, we used the capacity of the immunomodulatory peptides of arthropods with concomitant antimicrobial or antitumor activity. An SVM-based machine learning classifier identified short immunomodulatory sequences encrypted in 643 antimicrobial peptides from 55 foe-to-friend arthropods. The critical features involved in efficacy and safety were calculated. Finally, 76 safe immunomodulators were identified. Then, molecular docking and simulation studies defined the target of the most optimal peptide ligands among all human cell-surface TLRs. SPalf2-453 from a crab is a cell-penetrating immunoadjuvant with antiviral properties. The peptide interacts with the TLR1/2 heterodimer. SBsib-711 from a blackfly is a TLR4/MD2 ligand used as a cancer vaccine immunoadjuvant. In addition, SBsib-711 binds CD47 and PD-L1 on tumor cells, which is applicable in cancer immunotherapy as a checkpoint inhibitor. MRh4-679 from a shrimp is a broad-spectrum or universal immunoadjuvant with a putative Th1/Th2-balanced response. We also implemented a pathway enrichment analysis to define fingerprints or immunological signatures for further in vitro and in vivo immunogenicity and reactogenicity measurements. Conclusively, combinatorial machine learning, molecular docking, and simulation studies, as well as systems biology, open a new opportunity for the discovery and development of multifunctional prophylactic and therapeutic lead peptides.
Collapse
Affiliation(s)
- Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
| | - Zahra Saeidikia
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran;
| | - Hassan Seradj
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran;
| | - Abdolali Mohagheghzadeh
- Department of Phytopharmaceuticals, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran;
| |
Collapse
|
41
|
Mohammadian Farsani A, Mokhtari N, Nooraei S, Bahrulolum H, Akbari A, Farsani ZM, Khatami S, Ebadi MS, Ahmadian G. Lipid nanoparticles: The game-changer in CRISPR-Cas9 genome editing. Heliyon 2024; 10:e24606. [PMID: 38288017 PMCID: PMC10823087 DOI: 10.1016/j.heliyon.2024.e24606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/31/2024] Open
Abstract
The steady progress in genome editing, especially genome editing based on the use of clustered regularly interspaced short palindromic repeats (CRISPR) and programmable nucleases to make precise modifications to genetic material, has provided enormous opportunities to advance biomedical research and promote human health. However, limited transfection efficiency of CRISPR-Cas9 poses a substantial challenge, hindering its wide adoption for genetic modification. Recent advancements in nanoparticle technology, specifically lipid nanoparticles (LNPs), offer promising opportunities for targeted drug delivery. LNPs are becoming popular as a means of delivering therapeutics, including those based on nucleic acids and mRNA. Notably, certain LNPs, such as Polyethylene glycol-phospholipid-modified cationic lipid nanoparticles and solid lipid nanoparticles, exhibit remarkable potential for efficient CRISPR-Cas9 delivery as a gene editing instrument. This review will introduce the molecular mechanisms and diverse applications of the CRISPR/Cas9 gene editing system, current strategies for delivering CRISPR/Cas9-based tools, the advantage of LNPs for CRISPR-Cas9 delivery, an overview of strategies for overcoming off-target genome editing, and approaches for improving genome targeting and tissue targeting. We will also highlight current developments and recent clinical trials for the delivery of CRISPR/Cas9. Finally, future directions for overcoming the limitations and adaptation of this technology for clinical trials will be discussed.
Collapse
Affiliation(s)
- Arezoo Mohammadian Farsani
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Negin Mokhtari
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi Univesity, Tehran, Iran
| | - Saghi Nooraei
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Howra Bahrulolum
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Ali Akbari
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Zoheir Mohammadian Farsani
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Seyedmoein Khatami
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Mozhdeh sadat Ebadi
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Gholamreza Ahmadian
- Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
42
|
Zumuk CP, Jones MK, Navarro S, Gray DJ, You H. Transmission-Blocking Vaccines against Schistosomiasis Japonica. Int J Mol Sci 2024; 25:1707. [PMID: 38338980 PMCID: PMC10855202 DOI: 10.3390/ijms25031707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Control of schistosomiasis japonica, endemic in Asia, including the Philippines, China, and Indonesia, is extremely challenging. Schistosoma japonicum is a highly pathogenic helminth parasite, with disease arising predominantly from an immune reaction to entrapped parasite eggs in tissues. Females of this species can generate 1000-2200 eggs per day, which is about 3- to 15-fold greater than the egg output of other schistosome species. Bovines (water buffalo and cattle) are the predominant definitive hosts and are estimated to generate up to 90% of parasite eggs released into the environment in rural endemic areas where these hosts and humans are present. Here, we highlight the necessity of developing veterinary transmission-blocking vaccines for bovines to better control the disease and review potential vaccine candidates. We also point out that the approach to producing efficacious transmission-blocking animal-based vaccines before moving on to human vaccines is crucial. This will result in effective and feasible public health outcomes in agreement with the One Health concept to achieve optimum health for people, animals, and the environment. Indeed, incorporating a veterinary-based transmission vaccine, coupled with interventions such as human mass drug administration, improved sanitation and hygiene, health education, and snail control, would be invaluable to eliminating zoonotic schistosomiasis.
Collapse
Affiliation(s)
- Chika P. Zumuk
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - Malcolm K. Jones
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Severine Navarro
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Centre for Childhood Nutrition Research, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Darren J. Gray
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
| | - Hong You
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| |
Collapse
|
43
|
Gholap AD, Gupta J, Kamandar P, Bhowmik DD, Rojekar S, Faiyazuddin M, Hatvate NT, Mohanto S, Ahmed MG, Subramaniyan V, Kumarasamy V. Harnessing Nanovaccines for Effective Immunization─A Special Concern on COVID-19: Facts, Fidelity, and Future Prospective. ACS Biomater Sci Eng 2024; 10:271-297. [PMID: 38096426 DOI: 10.1021/acsbiomaterials.3c01247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Nanotechnology has emerged as a transformative pathway in vaccine research and delivery. Nanovaccines, encompassing lipid and nonlipid formulations, exhibit considerable advantages over traditional vaccine techniques, including enhanced antigen stability, heightened immunogenicity, targeted distribution, and the potential for codelivery with adjuvants or immune modulators. This review provides a comprehensive overview of the latest advancements and applications of lipid and non-lipid-based nanovaccines in current vaccination strategies for immunization. The review commences by outlining the fundamental concepts underlying lipid and nonlipid nanovaccine design before delving into the diverse components and production processes employed in their development. Subsequently, a comparative analysis of various nanocarriers is presented, elucidating their distinct physicochemical characteristics and impact on the immune response, along with preclinical and clinical studies. The discussion also highlights how nanotechnology enables the possibility of personalized and combined vaccination techniques, facilitating the creation of tailored nanovaccines to meet the individual patient needs. The ethical aspects concerning the use of nanovaccines, as well as potential safety concerns and public perception, are also addressed. The study underscores the gaps and challenges that must be overcome before adopting nanovaccines in clinical practice. This comprehensive analysis offers vital new insights into lipid and nonlipid nanovaccine status. It emphasizes the significance of continuous research, collaboration among interdisciplinary experts, and regulatory measures to fully unlock the potential of nanotechnology in enhancing immunization and ensuring a healthier, more resilient society.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Juhi Gupta
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Pallavi Kamandar
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Deblina D Bhowmik
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Md Faiyazuddin
- Department of Pharmaceutics, School of Pharmacy, Al-Karim University, Katihar 854106, Bihar, India
| | - Navnath T Hatvate
- Institute of Chemical Technology Mumbai, Marathwada Campus, Jalna 431213, Maharashtra, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru 575018, Karnataka, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru 575018, Karnataka, India
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
44
|
Malla R, Srilatha M, Farran B, Nagaraju GP. mRNA vaccines and their delivery strategies: A journey from infectious diseases to cancer. Mol Ther 2024; 32:13-31. [PMID: 37919901 PMCID: PMC10787123 DOI: 10.1016/j.ymthe.2023.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023] Open
Abstract
mRNA vaccines have evolved as promising cancer therapies. These vaccines can encode tumor-allied antigens, thus enabling personalized treatment approaches. They can also target cancer-specific mutations and overcome immune evasion mechanisms. They manipulate the body's cellular functions to produce antigens, elicit immune responses, and suppress tumors by overcoming limitations associated with specific histocompatibility leukocyte antigen molecules. However, successfully delivering mRNA into target cells destroys a crucial challenge. Viral and nonviral vectors (lipid nanoparticles and cationic liposomes) have shown great capacity in protecting mRNA from deterioration and assisting in cellular uptake. Cell-penetrating peptides, hydrogels, polymer-based nanoparticles, and dendrimers have been investigated to increase the delivery efficacy and immunogenicity of mRNA. This comprehensive review explores the landscape of mRNA vaccines and their delivery platforms for cancer, addressing design considerations, diverse delivery strategies, and recent advancements. Overall, this review contributes to the progress of mRNA vaccines as an innovative strategy for effective cancer treatment.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, AP, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati 517502, AP, India
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
45
|
Li M, Wang Y, Wang Y, Li R, Wang S, Ding P, Zhang G. Accurate location of two conserved linear epitopes of PEDV utilizing monoclonal antibodies induced by S1 protein nanoparticles. Int J Biol Macromol 2023; 253:127276. [PMID: 37804887 DOI: 10.1016/j.ijbiomac.2023.127276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/04/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Porcine Epidemic diarrhea virus (PEDV), which can result in severe vomiting, diarrhea, dehydration and death in newborn piglets, poses a great threat to the pig industry around the world. The S1 subunit of S protein is crucial for triggering neutralizing antibodies binding to the receptor. Based on the advantages of high immunogenicity and precise assembly of nanoparticles, the mi3 nanoparticles and truncated S1 protein were assembled by the SpyTag/SpyCatcher system and then expressed in HEK293F cells, whereafter high-efficiency monoclonal antibodies (mAbs) were produced and identified. The obtained five mAbs can bind to various genotypes of PEDV, including a mAb (12G) which can neutralize G1 and G2 genotypes of PEDV in vitro. By further identification of monoclonal antibody target sequences, 507FNDHSF512 and 553LFYNVTNSYG562 were first identified as B-cell linear epitopes, in which 553LFYNVTNSYG562 was a neutralizing epitope. Alanine scans identified the key amino acid sites of two epitopes. Moreover, the results of multiple sequence alignment analysis showed that these two epitopes were highly conserved in various subtype variants. In brief, these findings can serve as a basis for additional research of PEDV and prospective resources for the creation of later detection and diagnostic techniques.
Collapse
Affiliation(s)
- Minghui Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Yue Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Yanan Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Ruiqi Li
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Siqiao Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Peiyang Ding
- College of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Longhu Laboratory, Zhengzhou, China.
| | - Gaiping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China; College of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Longhu Laboratory, Zhengzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
46
|
Han R, Wang Y, Lu L. Sensitizing the Efficiency of ICIs by Neoantigen mRNA Vaccines for HCC Treatment. Pharmaceutics 2023; 16:59. [PMID: 38258070 PMCID: PMC10821464 DOI: 10.3390/pharmaceutics16010059] [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: 11/28/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
This study builds upon the groundbreaking mRNA vaccine Nobel Prize win in 2023 for COVID-19 prevention, paving the way for next-generation mRNA cancer vaccines to revolutionize immunotherapy. Despite the existing challenges, such as the presence of a suppressive tumor microenvironment and the identification of cancer-associated antigens, recent results from the KEYNOTE-942 trial have successfully demonstrated the effectiveness of mRNA-based cancer treatments, providing clinical evidence for the first time. This trial aimed to evaluate the efficacy and safety of combining immune checkpoint inhibitors with mRNA-based therapies in treating cancer. This advancement undeniably represents new hope for hepatocellular carcinoma (HCC) patients. However, progress in this field remains limited. In this article, we summarized the current state of applying immune checkpoint inhibitors (ICIs) combined with neoantigen mRNA vaccines. Additionally, we discussed potential targets for designing novel mRNA vaccines and potential mRNA vaccine delivery vehicles. The objective of this article is to inspire enthusiasm for the exploration of innovative therapeutic strategies that combine ICIs with neoantigen mRNA vaccines for HCC treatment and HCC prevention.
Collapse
Affiliation(s)
- Rui Han
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
- Department of Oncology, The First Hospital Affiliated to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
| | - Yuqian Wang
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT 06520-8034, USA
- School of Medicine, Center for Biomedical Data Science, New Haven, CT 06520-8034, USA
- Yale Cancer Center, Yale University, New Haven, CT 06520-8034, USA
| |
Collapse
|
47
|
Ma Y, Li S, Lin X, Chen Y. Bioinspired Spatiotemporal Management toward RNA Therapies. ACS NANO 2023; 17:24539-24563. [PMID: 38091941 DOI: 10.1021/acsnano.3c08219] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Ribonucleic acid (RNA)-based therapies have become an attractive topic in disease intervention, especially with some that have been approved by the FDA such as the mRNA COVID-19 vaccine (Comirnaty, Pfizer-BioNTech, and Spikevax, Moderna) and Patisiran (siRNA-based drug for liver delivery). However, extensive applications are still facing challenges in delivering highly negatively charged RNA to the targeted site. Therapeutic delivery strategies including RNA modifications, RNA conjugates, and RNA polyplexes and delivery platforms such as viral vectors, nanoparticle-based delivery platforms, and hydrogel-based delivery platforms as potential nucleic acid-releasing depots have been developed to enhance their cellular uptake and protect nucleic acid from being degraded by immune systems. Here, we review the growing number of viral vectors, nanoparticles, and hydrogel-based RNA delivery systems; describe RNA loading/release mechanism induced by environmental stimulations including light, heat, pH, or enzyme; discuss their physical or chemical interactions; and summarize the RNA therapeutics release period (temporal) and their target cells/organs (spatial). Finally, we describe current concerns, highlight current challenges and future perspectives of RNA-based delivery systems, and provide some possible research areas that provide opportunities for clinical translation of RNA delivery carriers.
Collapse
Affiliation(s)
- Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shiyao Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xin Lin
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27705, United States
| | - Yupeng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
48
|
Lin X, Wang Y, Fang K, Guo Z, Lin N, Li L. The application of nanoparticles in theranostic systems targeting breast cancer stem cells: current progress and future challenges. Stem Cell Res Ther 2023; 14:356. [PMID: 38072976 PMCID: PMC10712155 DOI: 10.1186/s13287-023-03584-1] [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: 01/08/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer (BC) is one of the diseases with the highest female mortality rates in the world and is closely related to breast cancer stem cells (BCSCs). Conventional breast cancer chemotherapy drugs target noncancer stem cells (non-CSCs), while cancer stem cells (CSCs) can still survive, which is an important reason for breast cancer drug resistance and local recurrence or distant metastasis. How to eradicate BCSCs while killing BCs is the key factor to improve the effect, and it is also an important scientific problem to be solved urgently. Therefore, targeted BCSC therapy has become a research hotspot. Interestingly, the emergence of nanotechnology provides a new idea for targeting BCSCs. This study summarizes the current application status of nanomaterials in targeting BCSCs, and attempts to construct a new type of lipid nanoparticle (LNP) that can target BCSCs through mRNA, providing a new idea for the treatment of BC.
Collapse
Affiliation(s)
- Xinyu Lin
- Oncology Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Ying Wang
- Oncology Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Kai Fang
- Oncology Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Zijian Guo
- Department of Oncological Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Nan Lin
- Qilu Hospital of Shandong University, Shandong, 250000, China
| | - Lihua Li
- Oncology Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China.
| |
Collapse
|
49
|
Liu X, Min Q, Song H, Yue A, Li Q, Zhou Q, Han W. Potentiating humoral and cellular immunity using a novel hybrid polymer-lipid nanoparticle adjuvant for HBsAg-VLP vaccine. J Nanobiotechnology 2023; 21:441. [PMID: 37993870 PMCID: PMC10666313 DOI: 10.1186/s12951-023-02116-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/16/2023] [Indexed: 11/24/2023] Open
Abstract
Aluminium adjuvants are commonly used in vaccines to stimulate the immune system, but they have limited ability to promote cellular immunity which is necessary for clearing viral infections like hepatitis B. Current adjuvants that do promote cellular immunity often have undesired side effects due to the immunostimulants they contain. In this study, a hybrid polymer lipid nanoparticle (HPLNP) was developed as an efficient adjuvant for the hepatitis B surface antigen (HBsAg) virus-like particle (VLP) vaccine to potentiate both humoral and cellular immunity. The HPLNP is composed of FDA approved polyethylene glycol-b-poly (L-lactic acid) (PEG-PLLA) polymer and cationic lipid 1, 2-dioleoyl-3-trimethylammonium-propane (DOTAP), and can be easily prepared by a one-step method. The cationic optimised vaccine formulation HBsAg/HPLNP (w/w = 1/600) can maximise the cell uptake of the antigen due to the electrostatic adsorption between the vaccine nanoparticle and the cell membrane of antigen-presenting cells. The HPLNP prolonged the retention of the antigen at the injection site and enhanced the lymph node drainage of antigen, resulting in a higher concentration of serum anti-HBsAg IgG compared to the HBsAg group or the HBsAg/Al group after the boost immunisation in mice. The HPLNP also promoted a strong Th1-driven immune response, as demonstrated by the significantly improved IgG2a/IgG1 ratio, increased production of IFN-γ, and activation of CD4 + and CD8 + T cells in the spleen and lymph nodes. Importantly, the HPLNP demonstrated no systemic toxicity during immunisation. The advantages of the HPLNP, including good biocompatibility, easy preparation, low cost, and its ability to enhance both humoral and cellular immune responses, suggest its suitability as an efficient adjuvant for protein-based vaccines such as HBsAg-VLP. These findings highlight the promising potential of the HPLNP as an HBV vaccine adjuvant, offering an alternative to aluminium adjuvants currently used in vaccines.
Collapse
Affiliation(s)
- Xuhan Liu
- Department of Emergency Medicine, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, No. 1098 Xueyuan Avenue, Shenzhen, 518000, Guangdong, China
| | - Qiuxia Min
- Department of Pharmacy, First People's Hospital of Yunnan Province, Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, 650034, Yunnan, China
| | - Huiping Song
- Department of Emergency Medicine, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, No. 1098 Xueyuan Avenue, Shenzhen, 518000, Guangdong, China
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Aochun Yue
- Department of Emergency Medicine, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, No. 1098 Xueyuan Avenue, Shenzhen, 518000, Guangdong, China
- Centre of Integrated Chinese and Western Medicine, School of Clinical Medicine, Qingdao University, Qingdao, China
| | - Qin Li
- Department of Emergency Medicine, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, No. 1098 Xueyuan Avenue, Shenzhen, 518000, Guangdong, China
| | - Qing Zhou
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wei Han
- Department of Emergency Medicine, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen University, No. 1098 Xueyuan Avenue, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
50
|
Prchalova E, Kohoutova Z, Knittelova K, Malinak D, Musilek K. Strategies for enhanced bioavailability of oxime reactivators in the central nervous system. Arch Toxicol 2023; 97:2839-2860. [PMID: 37642747 DOI: 10.1007/s00204-023-03587-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/10/2023] [Indexed: 08/31/2023]
Abstract
Oxime reactivators of acetylcholinesterase are commonly used to treat highly toxic organophosphate poisoning. They are effective nucleophiles that can restore the catalytic activity of acetylcholinesterase; however, their main limitation is the difficulty in crossing the blood-brain barrier (BBB) because of their strongly hydrophilic nature. Various approaches to overcome this limitation and enhance the bioavailability of oxime reactivators in the CNS have been evaluated; these include structural modifications, conjugation with molecules that have transporters in the BBB, bypassing the BBB through intranasal delivery, and inhibition of BBB efflux transporters. A promising approach is the use of nanoparticles (NPs) as the delivery systems. Studies using mesoporous silica nanomaterials, poly (L-lysine)-graft-poly(ethylene oxide) NPs, metallic organic frameworks, poly(lactic-co-glycolic acid) NPs, human serum albumin NPs, liposomes, solid lipid NPs, and cucurbiturils, have shown promising results. Some NPs are considered as nanoreactors for organophosphate detoxification; these combine bioscavengers with encapsulated oximes. This study provides an overview and critical discussion of the strategies used to enhance the bioavailability of oxime reactivators in the central nervous system.
Collapse
Affiliation(s)
- Eliska Prchalova
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Zuzana Kohoutova
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - Karolina Knittelova
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic
| | - David Malinak
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic.
- Biomedical Research Centre, University Hospital in Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic.
| | - Kamil Musilek
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03, Hradec Kralove, Czech Republic.
- Biomedical Research Centre, University Hospital in Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic.
| |
Collapse
|