1
|
Qiu N, Xu C, Zhang Z, Wang R, Wei X, Xie Y, Wang S, Lu D, Wang K, Xu S, Shen C, Su R, Cen B, Liu Y, Shen Y, Xu X. Autologous tumoral esterase-driven therapeutic polymers sequentially orchestrated antigen-induction, STING activation and anti-angiogenesis for systemic cancer immune therapy. Biomaterials 2025; 320:123260. [PMID: 40138966 DOI: 10.1016/j.biomaterials.2025.123260] [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: 10/30/2024] [Revised: 02/23/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025]
Abstract
Effective cancer immune therapy requires the orchestration of antigen induction, presentation and T-cell activation, further enhanced by anti-angiogenesis treatment; therefore, multiple therapeutics are generally used for such combination therapy. Herein, we report esterase-hydrolysable cationic polymers, N-[3-((4-acetoxy benzyl) oxy)-3-oxopropyl]-N-methyl-quaternized PEI (ERP) and poly{N-[2-(acryloyl-oxy) ethyl]-N-[p-acetyloxyphenyl]-N,N-dimethylammonium chloride} (PQDMA), capable of simultaneously inducing tumor cell immunogenic cell death (ICD) to release antigens, activating the cGAS-STING pathways of tumor macrophages and dendritic cells, and releasing antiangiogenic agent p-hydroxybenzyl alcohol (HBA). Thus, intratumoral injection of ERP or PQDMA systemically boosted the anti-cancer immunities and inhibited tumor angiogenesis in mouse hepatocellular carcinoma and melanoma bilateral tumor models, leading to more effective tumor growth inhibition of both treated and abscopal untreated tumors than ICD alone induced by mitoxantrone and control cationic polymers. Further study using gene knockout mice and transcriptome sequencing analysis confirmed the involvement of cGAS-STING and type I IFN signaling pathways. This work demonstrates ERP and PQDMA as the first examples of inherent therapeutic polymers, accomplishing systemic tumor inhibition without combining other therapeutic agents.
Collapse
Affiliation(s)
- Nasha Qiu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China.
| | - Chang Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Zhen Zhang
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education of China, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Rui Wang
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Yangla Xie
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Di Lu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310059, China
| | - Kai Wang
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310059, China
| | - Shengjun Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Chenchen Shen
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Renyi Su
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Beini Cen
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Yanpeng Liu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Youqing Shen
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education of China, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310059, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Dalla Pietà A, Genova B, Penna A, Sinigaglia A, Vogiatzis S, Barzon L, Pagliari M, Bonfante F, Torrigiani F, Sofia T, Verin R, Tosi A, Carpanese D, Sommaggio R, Barbieri V, Dalla Santa S, Zuccolotto G, Grigoletto A, Pasut G, Rosato A. On the adjuvanticity of hyaluronan: The case of a SARS-CoV-2 vaccine. J Control Release 2025; 382:113674. [PMID: 40164435 DOI: 10.1016/j.jconrel.2025.113674] [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: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Vaccines based on mRNA have been fundamental in facing the COVID-19 pandemic, however, they still raise concerns about stability and long-term efficacy. Thus, protein-based vaccines remain valid options and hence the study of effective adjuvants is crucial. Here, we developed a COVID-19 vaccine based on the receptor-binding domain (RBD) of SARS-CoV-2 Spike protein, which is covalently conjugated to the natural polymer hyaluronan (HA) that acts as an immunological adjuvant. Vaccination of K18-hACE2 mice with HA-RBD was well tolerated, and elicited high and sustained titres of RBD-binding antibodies and SARS-CoV-2-neutralizing antibodies, without the addition of other immunostimulatory compounds. Most importantly, HA-RBD vaccination conferred long-term protection to K18-hACE2 mice after challenge with SARS-CoV-2, also in the case of two consequent infections driven by different variants. These findings demonstrate the efficacy of HA-based vaccination against COVID-19 disease, and support the promising use of HA as an efficient and well tolerated adjuvant.
Collapse
Affiliation(s)
- Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy
| | - Beatrice Genova
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy
| | - Alessandro Penna
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Alessandro Sinigaglia
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Stefania Vogiatzis
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Matteo Pagliari
- Department of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università 10, 35020 Legnaro, PD, Italy
| | - Francesco Bonfante
- Department of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università 10, 35020 Legnaro, PD, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, 35020 Legnaro, PD, Italy
| | - Tomasoni Sofia
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, 35020 Legnaro, PD, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, 35020 Legnaro, PD, Italy
| | - Anna Tosi
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Debora Carpanese
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Roberta Sommaggio
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy; Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Vito Barbieri
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy
| | - Silvia Dalla Santa
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Antonella Grigoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Francesco Marzolo 5, 35131 Padua, Italy
| | - Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Francesco Marzolo 5, 35131 Padua, Italy.
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy; Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy.
| |
Collapse
|
3
|
Zhang H, Xu X, Li S, Huang H, Zhang K, Li W, Wang X, Yang J, Yin X, Qu C, Ni J, Dong X. Advances in nanoplatform-based multimodal combination therapy activating STING pathway for enhanced anti-tumor immunotherapy. Colloids Surf B Biointerfaces 2025; 250:114573. [PMID: 39983453 DOI: 10.1016/j.colsurfb.2025.114573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/24/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Activation of the cyclic GMP-AMP synthase(cGAS)-stimulator of interferon genes (STING) has great potential to promote antitumor immunity. As a major effector of the cell to sense and respond to the aberrant presence of cytoplasmic double-stranded DNA (dsDNA), inducing the expression and secretion of type I interferons (IFN) and STING, cGAS-STING signaling pathway establishes an effective natural immune response, which is one of the fundamental mechanisms of host defense in organisms. In addition to the release of heterologous DNA due to pathogen invasion and replication, mitochondrial damage and massive cell death can also cause abnormal leakage of the body's own dsDNA, which is then recognized by the DNA receptor cGAS and activates the cGAS-STING signaling pathway. However, small molecule STING agonists suffer from rapid excretion, low bioavailability, non-specificity and adverse effects, which limits their therapeutic efficacy and in vivo application. Various types of nano-delivery systems, on the other hand, make use of the different unique structures and surface modifications of nanoparticles to circumvent the defects of small molecule STING agonists such as fast metabolism and low bioavailability. Also, the nanoparticles are precisely directed to the focal site, with their own appropriate particle size combined with the characteristics of passive or active targeting. Herein, combined with the cGAS-STING pathway to activate the immune system and kill tumor tissues directly or indirectly, which help maximize the use of the functions of chemotherapy, photothermal therapy(PTT), chemodynamic therapy(CDT), and radiotherapy(RT). In this review, we will discuss the mechanism of action of the cGAS-STING pathway and introduce nanoparticle-mediated tumor combination therapy based on the STING pathway. Collectively, the effective multimodal nanoplatform, which can activate cGAS-STING pathway for enhanced anti-tumor immunotherapy, has promising avenue clinical applications for cancer treatment.
Collapse
Affiliation(s)
- Huizhong Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaohan Xu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shiman Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huating Huang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ke Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenjing Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinzhu Wang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingwen Yang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingbin Yin
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Changhai Qu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xiaoxv Dong
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
4
|
Kwon KW, Choi E, Kim H, Kim HW, Choi S, Lee S, Ha SJ, Shin SJ. Adjunctive beneficial effect of c-di-GMP, a STING agonist, in enhancing protective efficacy of TLR4-adjuvanted tuberculosis subunit vaccine formulations. J Biomed Sci 2025; 32:52. [PMID: 40414893 DOI: 10.1186/s12929-025-01144-8] [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: 10/29/2024] [Accepted: 05/06/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Effective subunit vaccine development requires selecting appropriate adjuvant formulations to trigger desired adaptive immune responses. This study explores the immunogenicity and tuberculosis (TB) vaccine potential of antigens (Ags) combined with Toll-like receptor 4 (TLR4) adjuvants and a stimulator of interferon genes (STING) agonist. METHODS In this work, we investigated the combination of Ags with TLR4 adjuvants (monophosphoryl lipid A / dimethyldioctadecylammonium bromide; MPL/DDA or glucopyranosyl lipid adjuvant-stable emulsion; GLA-SE) and a STING agonist, c-di-GMP (CDG). Mice were immunized three times by intramuscular injections at 3-week intervals. The effects of integrating Ags in these adjuvant formulations on the immune response were evaluated, focusing on the generation of Th1-biased, polyfunctional Ag-specific CD4+ T cells and their localization in the lung and spleen. To assess protection, immunized mice were aerogenically challenged with either conventional or ultra-low doses of Mycobacterium tuberculosis (Mtb) 4 weeks after the last immunization. Subsequently, bacterial load and pulmonary inflammation were assessed. RESULTS Integrating ESAT6 Ag in TLR4 and CDG adjuvant formulations remarkably boosted Th1-biased, polyfunctional ESAT6-specific CD4+ T cells in the lungs and spleen, providing durable protection against Mtb infection. The inclusion of CDG promoted mucosal localization of ESAT6-specific CD4+ T cells resembling resident memory phenotypes in the lung parenchyma and increased Ag-specific CD4+ T cells in lung vasculature. Immunization with another vaccine Ag candidate, Ag85B, in GLA-SE plus CDG similarly increased Ag85B-specific CD4+ T cells in the spleen and both lung compartments. Following ultra-low dose Mtb challenge, ESAT6 or Ag85B/GLA-SE/CDG immunizations significantly reduced bacterial loads compared to non-, Bacillus Calmette-Guérin (BCG)-, and ESAT6 or Ag85B/GLA-SE-immunized groups. Importantly, the inclusion of CDG decreased killer cell lectin-like receptor subfamily G member 1 (KLRG1) expression among Ag-specific CD4+ T cells in the lung, correlating with enhanced lung-homing evidenced by expanded lung parenchyma Ag-specific CD4+ T cells, including less-differentiated Th1 cells. CONCLUSIONS This study highlights that CDG, when used in combination with TLR4 adjuvants, enhances long-term protective immunity, offering a promising strategy for subunit TB vaccine development.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology and Convergence of Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Eunsol Choi
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hagyu Kim
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hyeong Woo Kim
- Department of Microbiology and Convergence of Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Sangwon Choi
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Seunghyun Lee
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, 03722, South Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
5
|
Yıldırım M, Poyraz S, Acet Ö, Acet BÖ, Karakoç V, Odabaşı M. Chitosan hydrogels: Versatile platforms for drug delivery in cancer treatment, wound dressing, and 3D bioprinting applications. Int J Biol Macromol 2025; 314:144367. [PMID: 40398793 DOI: 10.1016/j.ijbiomac.2025.144367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 05/04/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
Chitosan, derived from chitin, is frequently employed in various applications, including hydrogels. In cancer treatment, chitosan serves as a drug carrier, enhancing drug bioavailability while reducing side effects. Additionally, its inherent antibacterial properties and ability to maintain a moist environment facilitate faster wound healing. Its capacity for controlled drug release also ensures prolonged delivery of therapeutic agents. Furthermore, its biocompatibility and biodegradability present substantial advantages. Beyond conventional methods, chitosan is now being utilized as a bioink in 3D printing technologies. This innovation enables personalized treatments, leveraging the advantages of chitosan. However, certain challenges must be addressed to ensure the proper application of this technology. This review not only provides comprehensive insights into the synthesis and biomedical applications of chitosan hydrogels but also summarizes recent studies from the past five years, focusing on their roles in wound healing, cancer treatment, and 3D printing applications.
Collapse
Affiliation(s)
- Metin Yıldırım
- Department of Biochemistry, Faculty of Pharmacy, Harran University, Şanlıurfa, Türkiye.
| | - Samet Poyraz
- Department of Analytical Chemistry, Faculty of Pharmacy, Ağrı İbrahim Çeçen University, Ağrı, Türkiye
| | - Ömür Acet
- Department of Pharmacy Services, Vocational School of Health Services, Tarsus University, Mersin, Türkiye
| | - Burcu Önal Acet
- Department of Chemistry, Faculty of Arts and Science, Aksaray University, Aksaray, Türkiye
| | - Veyis Karakoç
- Department of Chemistry, Faculty of Science, Cankiri Karatekin University, Cankiri, Türkiye
| | - Mehmet Odabaşı
- Department of Chemistry, Faculty of Arts and Science, Aksaray University, Aksaray, Türkiye
| |
Collapse
|
6
|
Chenuet P, Mellier M, Messaoud-Nacer Y, Culerier E, Marquant Q, Fauconnier L, Rouxel N, Ledru A, Rose S, Ryffel B, Apetoh L, Quesniaux VF, Togbe D. Birch pollen allergen-induced dsDNA release activates cGAS-STING signaling and type 2 immune response in mice. iScience 2025; 28:112324. [PMID: 40276777 PMCID: PMC12018559 DOI: 10.1016/j.isci.2025.112324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/26/2024] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Detecting cytoplasmic or extracellular DNA from host or pathogen origin by DNA sensor cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) triggers immune responses with secretion of type I interferons and inflammatory cytokines. However, STING agonists function as type-2 adjuvant promoting allergic asthma. Here, we asked how cGAS/STING signaling pathway influences allergen-induced type-2 immune responses in models of allergic airway diseases induced by birch pollen extract, house dust mite, or ovalbumin plus Alum. We report increased extracellular dsDNA in the airways, together with cGAS and STING gene expression, following allergen challenge in these models, correlating dsDNA and type-2 cytokine IL-4, IL-5, and IL-13 release. Allergen-induced type-2 immune responses were reduced in cGAS- or STING-deficient mice. Further, blocking cGAS function with the specific inhibitor RU.521 protected mice from birch pollen allergen-induced airway inflammation and type-2 immune responses. Thus, DNA sensing by cGAS contributes to type-2 immune responses and may represent a therapeutic target for allergic lung inflammation.
Collapse
Affiliation(s)
| | - Manon Mellier
- Artimmune SAS, 13 Avenue Buffon, 45100 Orléans, France
| | - Yasmine Messaoud-Nacer
- Laboratory of Immuno-Neuro Modulation (INEM), UMR 7355 CNRS and University of Orleans, 3B rue de la Ferollerie, 45071 Orleans-Cedex, France
| | - Elodie Culerier
- Laboratory of Immuno-Neuro Modulation (INEM), UMR 7355 CNRS and University of Orleans, 3B rue de la Ferollerie, 45071 Orleans-Cedex, France
| | - Quentin Marquant
- Laboratory of Immuno-Neuro Modulation (INEM), UMR 7355 CNRS and University of Orleans, 3B rue de la Ferollerie, 45071 Orleans-Cedex, France
| | | | | | - Aurélie Ledru
- Artimmune SAS, 13 Avenue Buffon, 45100 Orléans, France
| | - Stéphanie Rose
- Laboratory of Immuno-Neuro Modulation (INEM), UMR 7355 CNRS and University of Orleans, 3B rue de la Ferollerie, 45071 Orleans-Cedex, France
| | - Bernhard Ryffel
- Artimmune SAS, 13 Avenue Buffon, 45100 Orléans, France
- Laboratory of Immuno-Neuro Modulation (INEM), UMR 7355 CNRS and University of Orleans, 3B rue de la Ferollerie, 45071 Orleans-Cedex, France
| | - Lionel Apetoh
- Brown Center for Immunotherapy, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Valérie F.J. Quesniaux
- Laboratory of Immuno-Neuro Modulation (INEM), UMR 7355 CNRS and University of Orleans, 3B rue de la Ferollerie, 45071 Orleans-Cedex, France
| | - Dieudonnée Togbe
- Laboratory of Immuno-Neuro Modulation (INEM), UMR 7355 CNRS and University of Orleans, 3B rue de la Ferollerie, 45071 Orleans-Cedex, France
- University of Orleans, 45000 Orleans, France
| |
Collapse
|
7
|
Mendoza-Guevara CC, Martinez-Escobar A, Ramos-Godínez MDP, Muñoz-Medina JE, Ramon-Gallegos E. Efficient Delivery of SARS-CoV-2 Plasmid DNA in HEK-293T Cells Using Chitosan Nanoparticles. Pharmaceuticals (Basel) 2025; 18:683. [PMID: 40430502 DOI: 10.3390/ph18050683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Gene therapy has emerged as a promising strategy for treating a wide range of diseases. However, a major challenge remains in developing efficient and safe delivery systems for genetic material. Nanoparticles, particularly chitosan nanoparticles (CNPs), have gained significant attention as a potential solution. This study focuses on designing a SARS-CoV-2 plasmid DNA (pDNA) conjugated with CNPs and evaluating its in vitro delivery efficiency. Methods: The Omicron Spike DNA sequence was inserted into the pIRES2-eGFP expression vector, and CNPs were synthesized with optimized physicochemical properties to enhance stability, cellular uptake, and transfection efficiency. The conjugate was characterized using UV-Vis, FT-IR, DLS, and TEM techniques. Transfection efficiency was assessed and compared to the commercially available TurboFect reagent as a control. Results: CNPs-pDNA polyplexes with an average size of 159.0 ± 33.1 nm (TEM), a zeta potential of +19.7 ± 0.3 mV, and 100% ± 0.0 encapsulation efficiency were developed as a non-viral delivery system. CNPs efficiently serve as a delivery vehicle for the constructed pDNA without altering cell morphology, achieving transfection efficiencies of 62-74%, compared to 55-70% for TurboFect. Furthermore, RT-qPCR confirmed the expression of Spike mRNA, and Western blot assays validated the expression of Spike protein. Notably, Spike protein expression from CNPs was found to be two-fold higher than the control at 96 h post-transfection. Conclusions: These findings suggest that CNPs are a promising and versatile platform for delivering genetic material. Importantly, this study highlights the intrinsic properties of chitosan, without the use of additional ligands, as a key factor in achieving efficient gene delivery.
Collapse
Affiliation(s)
- Citlali Cecilia Mendoza-Guevara
- Laboratorio de Citopatología Ambiental, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Ciudad de México 07738, Mexico
| | - Alejandro Martinez-Escobar
- Laboratorio de Citopatología Ambiental, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Ciudad de México 07738, Mexico
| | | | - José Esteban Muñoz-Medina
- Banco de Muestras de la CCILE, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México 07760, Mexico
| | - Eva Ramon-Gallegos
- Laboratorio de Citopatología Ambiental, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Ciudad de México 07738, Mexico
| |
Collapse
|
8
|
Duymaz D, Kebabci AO, Kizilel S. Harnessing the immunomodulatory potential of chitosan and its derivatives for advanced biomedical applications. Int J Biol Macromol 2025; 307:142055. [PMID: 40090654 DOI: 10.1016/j.ijbiomac.2025.142055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
The success of biomaterial applications in medicine, particularly in tissue engineering, relies on achieving a balance between promoting tissue regeneration and controlling the immune response. Due to its natural origin, high biocompatibility, and versatility, chitosan has emerged as a promising biomaterial especially for immunomodulation purposes. Immunomodulation, refers to the deliberate alteration of the immune system's activity to achieve a desired therapeutic effect either by enhancing or suppressing the function of specific immune cells, signaling pathways, or cytokine production. This modulation opens up the unlimited possibilities for the use of biomaterials, especially about the use of natural polymers such as chitosan. Although numerous chitosan-based immunoregulatory strategies have been demonstrated over the past two decades, the lack of in-depth exploration hinders the full potential of strategies that include chitosan and its derivatives in biomedical applications. Thus, in this review, the possible immunomodulatory effects of chitosan, chitosan derivatives and their potential combined with various agents and therapies are investigated in detail. Moreover, this report includes agents for localized immune response control, chitosan-based strategies with complementary immunomodulatory properties to create synergistic effects that will influence the success of cell therapies for enhanced tissue acceptance and regeneration. Finally, the challenges and outlook of chitosan-based therapies as a powerful tool for improving immunomodulatory applications are discussed for paving the way for further studies.
Collapse
Affiliation(s)
- Doğukan Duymaz
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Aybaran O Kebabci
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Seda Kizilel
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye.
| |
Collapse
|
9
|
Ahmed M, Kurungottu P, Swetha K, Atla S, Ashok N, Nagamalleswari E, Bonam SR, Sahu BD, Kurapati R. Role of NLRP3 inflammasome in nanoparticle adjuvant-mediated immune response. Biomater Sci 2025; 13:2164-2178. [PMID: 38867716 DOI: 10.1039/d4bm00439f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is pivotal in orchestrating the immune response induced by nanoparticle adjuvants. Understanding the intricate mechanisms underlying the activation of NLRP3 inflammasome by these adjuvants is crucial for deciphering their immunomodulatory properties. This review explores the involvement of the NLRP3 inflammasome in mediating immune responses triggered by nanoparticle adjuvants. It delves into the signaling pathways and cellular mechanisms involved in NLRP3 activation, highlighting its significance in modulating the efficacy and safety of nanoparticle-based adjuvants. A comprehensive grasp of the interplay between NLRP3 inflammasome and nanoparticle adjuvants holds promise for optimizing vaccine design and advancing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Momitul Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Pavithra Kurungottu
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - K Swetha
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Sandeep Atla
- Texas A&M Drug Discovery Center, Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | - Nivethitha Ashok
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Easa Nagamalleswari
- MTCC and Gene Bank, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| |
Collapse
|
10
|
Wang X, He Q, Wang L, Li C, Zhang W, Rong Z, Yin Q, Zhao Y. Acid responsive molybdenum (Mo)-based nanoparticles inhibit the cGAS-STING signaling pathway for sepsis therapy. Biomater Sci 2025; 13:2410-2421. [PMID: 40130269 DOI: 10.1039/d5bm00007f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Sepsis, an inflammatory disease caused by bacterial infection, has become a global public health crisis. Excessive reactive oxygen species (ROS) in sepsis patients act as the primary trigger for activating intracellular immune pathways, ultimately leading to multiple organ dysfunction syndrome. The overexpression of acidic metabolites and ROS, characteristic of the infected microenvironment, significantly impedes sepsis treatment. Cyclic GMP-AMP synthase (cGAS), a cytosolic DNA sensor, plays a key role in inflammatory diseases. The detrimental effects of STING in sepsis have been well documented. Here, we developed a pH-responsive nanotherapy platform (DMSNM@C-178/PAA) that combines ROS scavenging with cGAS-STING pathway inhibition for anti-inflammatory therapy. This nanoparticle is selectively released in the infected microenvironment, where reduced molybdenum-based polyoxometalates (Mo-POM) efficiently neutralize toxic ROS in vivo, while C-178 selectively inhibits the cGAS-STING pathway, thereby attenuating the inflammatory response and preventing organ deterioration. In vitro and in vivo studies demonstrate that DMSNM@C-178/PAA treats sepsis by eliminating excess ROS and modulating autoimmune dysfunction via the cGAS-STING pathway, providing a novel therapeutic strategy for sepsis management.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Qingbin He
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Lining Wang
- Department of Oncology No.1, Rushan People's Hospital, Rushan 264500, Shandong, China
| | - Chengzhilin Li
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Wenyu Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Zhonghou Rong
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yingchun Zhao
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| |
Collapse
|
11
|
Qiao Y, Wei L, Su Y, Tan Q, Yang X, Li S. Nanoparticle-Based Strategies to Enhance the Efficacy of STING Activators in Cancer Immunotherapy. Int J Nanomedicine 2025; 20:5429-5456. [PMID: 40308645 PMCID: PMC12042967 DOI: 10.2147/ijn.s515893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a critical role in triggering innate and adaptive immune responses through type I interferon activation and immune cell recruitment, holding significant promise for cancer therapy. While STING activators targeting this pathway have been developed, their clinical application is hindered by challenges such as poor membrane permeability, rapid degradation, suboptimal pharmacokinetics, off-target biodistribution, and toxicity. Nanoparticle-based delivery systems offer a promising solution by enhancing the stability, circulation time, tumor accumulation, and intracellular release of STING activators. Furthermore, combining nanoparticle-delivered STING activators with radiotherapy, chemotherapy, phototherapy, and other immunotherapies enables synergistic antitumor effects through multimodal mechanisms, addressing resistance to monotherapies and reducing risks of recurrence and metastasis. This review outlines the immunomodulatory mechanisms of the cGAS-STING pathway, surveys current STING-targeted activators, and comprehensively discusses recent advances in nanoparticle-mediated delivery strategies for STING activation. Additionally, we explore combinatorial approaches that integrate STING-targeted nanotherapies with conventional and emerging treatments. Finally, we highlight the current status, prospects, and challenges of nanoparticle-based STING activation for cancer immunotherapy.
Collapse
Affiliation(s)
- Yi Qiao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Lingyu Wei
- Department of Gynecologic Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, People’s Republic of China
| | - Yinjie Su
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Qinyuan Tan
- Department of Urology, The People’s Hospital of Jimo, Qingdao, People’s Republic of China
| | - Xuecheng Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Shengxian Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
12
|
Li Z, Chen P, Qu A, Sun M, Xu L, Xu C, Hu S, Kuang H. Opportunities and Challenges for Nanomaterials as Vaccine Adjuvants. SMALL METHODS 2025:e2402059. [PMID: 40277301 DOI: 10.1002/smtd.202402059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/29/2025] [Indexed: 04/26/2025]
Abstract
Adjuvants, as a critical component of vaccines, are capable of eliciting more robust and sustained immune responses. Nanomaterials have shown unique advantages and broad application prospects in adjuvant development due to their high adjustability and distinctive physicochemical properties. This review focuses on nanoadjuvants and their immunological mechanisms. First, various types of adjuvants are introduced with an emphasis on metal and metal oxide nanoparticles, coordination polymers, liposomes, polymer nanoparticles, and other inorganic nanoparticles that can serve as vaccine adjuvants. Second, this review describes the current status of the clinical applications of nanoadjuvants. Next, the mechanisms of action for nanoadjuvants have been thoroughly elucidated, including the depot effect, NLRP3 inflammasome activation, targeting C-type lectin receptors, activation of toll-like receptors, and activation of the cGAS-STING signaling pathway. Finally, the challenges and opportunities associated with the development of nanoadjuvants have also been addressed.
Collapse
Affiliation(s)
- Zongda Li
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Panpan Chen
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aihua Qu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Shudong Hu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
13
|
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
|
14
|
Lian J, Men Z, Xu X, Li Y, Li J, Wang W, Yao T, Li Y, Qu Y, Feng Y, Wang S. Effects of folic acid supplementation before conception on innate immunity and anti-HBs levels of offspring born to HBsAg-positive mothers. Front Nutr 2025; 12:1526053. [PMID: 40336960 PMCID: PMC12055552 DOI: 10.3389/fnut.2025.1526053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/24/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction Folic acid has been associated with fetal development, especially in fetal immunity. Therefore, limited evidence regarding the effects of different folic acid supplementation of hepatitis B surface antigen (HBsAg) positive mothers in innate immunity in offspring. Herein, this study aimed to explore the association between folic acid supplementation and the innate immunity of neonates and the immunological efficacy of hepatitis B vaccine (HepB), which may provide insights that could inform pre-pregnancy health management in HBsAg-positive mothers. Materials and methods It is an ambispective cohort study with 293 pairs of HBsAg-positive mothers-offspring in Taiyuan, Shanxi Province, China. Mothers were classified into three groups according to the time of starting folic acid supplementation, non-supplementation group, pre-pregnancy group and post-pregnancy supplementation group. Immunological indexes such as immune cells proportion and innate immune mediators in cord blood and anti-HBs in infants were measured. Differences in immunological indexes were analyzed by One-Way ANOVA test. Univariate and multivariate analyses were performed for factors associated with abnormal immunological indexes and potential confounders were adjusted. Results The preconception folic acid group showed a significantly higher expression levels of STING (P = 0.005) and pNF-κB (P = 0.010) in cord blood along with higher anti-HBs titres (P = 0.006), when compared to both non-supplementation group and post-pregnancy supplementation group. Higher anti-HBs levels indicate a stronger immune response to HepB and may enhance protection against HBV infection during early life. Infants in the high pNF-κB expression group exhibited a significantly elevated seropositive rate of HepB compared to those in the low pNF-κB expression group (P = 0.037). There were no mediation effects and no moderation effects in this study, potentially due to the direct influence of folic acid supplementation on immune responses or the limited sample size. Discussion In conclusion, our findings demonstrate that preconception folic acid supplementation may enhance HepB vaccine responsiveness in infants of HBsAg-positive mothers. Meanwhile, high pNF-κB expression in cord blood can increase seropositive rates in infants. This discovery has significant public health implications, as it may provide a simple and accessible intervention to improve vaccination outcomes and reduce HBV transmission in endemic regions.
Collapse
Affiliation(s)
- Jia Lian
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Reverse Etiology Research Center Academician Workstation, Shanxi Medical University, Taiyuan, China
| | - Zhaoyue Men
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Reverse Etiology Research Center Academician Workstation, Shanxi Medical University, Taiyuan, China
| | - Xiuyang Xu
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Reverse Etiology Research Center Academician Workstation, Shanxi Medical University, Taiyuan, China
| | - Yandi Li
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Reverse Etiology Research Center Academician Workstation, Shanxi Medical University, Taiyuan, China
| | - Jinbo Li
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Reverse Etiology Research Center Academician Workstation, Shanxi Medical University, Taiyuan, China
| | - Weigang Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Reverse Etiology Research Center Academician Workstation, Shanxi Medical University, Taiyuan, China
| | - Tian Yao
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuping Li
- Department of Obstetrics and Gynaecology, The Third People's Hospital of Taiyuan, Taiyuan, China
| | - Yiqun Qu
- Department of Obstetrics and Gynaecology, The Third People's Hospital of Taiyuan, Taiyuan, China
| | - Yongliang Feng
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Reverse Etiology Research Center Academician Workstation, Shanxi Medical University, Taiyuan, China
| | - Suping Wang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
- Center of Clinical Epidemiology and Evidence Based Medicine, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Reverse Etiology Research Center Academician Workstation, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
15
|
Obeidat WM, Lahlouh IK. Chitosan Nanoparticles: Approaches to Preparation, Key Properties, Drug Delivery Systems, and Developments in Therapeutic Efficacy. AAPS PharmSciTech 2025; 26:108. [PMID: 40244367 DOI: 10.1208/s12249-025-03100-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
The integration of nanotechnology into drug delivery systems holds great promise for enhancing pharmaceutical effectiveness. This approach enables precise targeting, controlled release, improved patient compliance, reduced side effects, and increased bioavailability. Nanoparticles are vital for transporting biomolecules-such as proteins, enzymes, genes, and vaccines-through various administration routes, including oral, intranasal, vaginal, buccal, and pulmonary. Among biodegradable polymers, chitosan, a linear polysaccharide derived from chitin, stands out due to its biocompatibility, safety, biodegradability, mucoadhesive properties, and ability to enhance permeation. Its cationic nature supports strong molecular interactions and provides antimicrobial, anti-inflammatory, and hemostatic benefits. However, its solubility, influenced by pH and ionic sensitivity, poses challenges requiring effective solutions. This review explores chitosan, its modified derivatives and chitosan nanoparticles mainly, focusing on nanoparticles physicochemical properties, drug release mechanisms, preparation methods, and factors affecting their mean hydrodynamic diameter (particle size). It highlights their application in drug delivery systems and disease treatments across various routes. Key considerations include drug loading capacity, zeta potential, and stability, alongside the impact of molecular weight, degree of deacetylation, and drug solubility on nanoparticle properties. Recent advancements and studies underscore chitosan's potential, emphasizing its modified derivatives'versatility in improving therapeutic outcomes.
Collapse
Affiliation(s)
- Wasfy M Obeidat
- Jordan University of Science and Technology, 3030, Irbid, 22110, Jordan.
| | - Ishraq K Lahlouh
- Jordan University of Science and Technology, 3030, Irbid, 22110, Jordan
| |
Collapse
|
16
|
Liu C, Liu M, Li X, Hu Y, Zhang L, You FM, Fan G, Ge Y. Unique advantages and applications of polysaccharide microneedles as drug delivery materials and in treatment of skin diseases. NANOSCALE ADVANCES 2025:d4na01083c. [PMID: 40417161 PMCID: PMC12096177 DOI: 10.1039/d4na01083c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/11/2025] [Indexed: 05/27/2025]
Abstract
Owing to its non-invasive nature, painless drug delivery, and controlled drug loading capacity, the microneedle (MN) technology has recently garnered significant attention in clinical practice. For instance, it has been pervasively employed as an innovative transdermal delivery method in skin disease therapy. However, traditional MN techniques have been associated with challenges regarding biocompatibility, biodegradability, and drug release precision, limiting their clinical efficacy and increasing the risk of side effects resulting from uneven drug distribution. To address these issues, polysaccharide materials have been proposed as viable alternatives to be used in MN technologies. In addition to their excellent biocompatibility and biodegradability, polysaccharide materials such as alginate, chitosan, and Hyaluronic Acid (HA), among other Traditional Chinese Medicine (TCM)-extracted polysaccharides (such as Bletilla and notoginseng), could also exert anti-inflammatory and antibacterial effects, promoting tissue regeneration. These attributes enable polysaccharide-based MNs to improve the local drug concentration, reduce systemic side effects, minimize patient discomfort, and lower treatment risks, making them particularly suitable for treating skin conditions such as eczema, psoriasis, and acne. This article systematically reviews the properties of various polysaccharide materials, as well as the preparation methods of polysaccharide-based MNs and their therapeutic effects as reported in animal models and clinical trials. Our findings could lay a solid theoretical foundation for developing polysaccharide-based MN technologies and fostering their widespread clinical application.
Collapse
Affiliation(s)
- Chao Liu
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu 610072 China
- Chengdu University of Traditional Chinese Medicine Chengdu 611137 China
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine China
| | - Meng Liu
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu 610072 China
- Chengdu University of Traditional Chinese Medicine Chengdu 611137 China
| | - Xin Li
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu 610072 China
- Chengdu University of Traditional Chinese Medicine Chengdu 611137 China
| | - Yimei Hu
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu 610072 China
- Chengdu University of Traditional Chinese Medicine Chengdu 611137 China
| | - Lingling Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu 610072 China
| | - Feng-Ming You
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu 610072 China
- Chengdu University of Traditional Chinese Medicine Chengdu 611137 China
| | - Gang Fan
- Chengdu University of Traditional Chinese Medicine Chengdu 611137 China
| | - Yiman Ge
- Hospital of Chengdu University of Traditional Chinese Medicine Chengdu 610072 China
- Chengdu University of Traditional Chinese Medicine Chengdu 611137 China
| |
Collapse
|
17
|
Ling X, Dong Z, He J, Chen D, He D, Guo R, He Q, Li M. Advances in Polymer-Based Self-Adjuvanted Nanovaccines. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409021. [PMID: 40079071 DOI: 10.1002/smll.202409021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/22/2025] [Indexed: 03/14/2025]
Abstract
Nanovaccines, as a new generation of vaccines, have garnered significant interest due to their exceptional potential in enhancing disease prevention and treatment. Their unique features, such as high stability, antigens protection, prolonged retention, and targeted delivery to lymph nodes, immune cells, and tumors, set them apart as promising candidates in the field of immunotherapy. Polymers, with their superior degradability, capacity to mimic pathogen characteristics, and surface functionality that facilitates modifications, serve as ideal carriers for vaccine components. Polymer-based self-adjuvanted nanovaccines have the remarkable ability to augment immune responses. The inherent adjuvant-like properties of polymers themselves offer a pathway toward more efficient exploitation of nanomaterials and the optimization of nanovaccines. This review article aims to summarize the categorization of polymers and elucidate their mechanisms of action as adjuvants. Additionally, it delves into the advantages and limitations of polymer-based self-adjuvanted nanovaccines in disease management and prevention, providing valuable insights for their design and application. This comprehensive analysis could contribute to the development of more effective and tailored nanovaccines for a wide range of diseases.
Collapse
Affiliation(s)
- Xiaoli Ling
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Ziyan Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Jiao He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Dong Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Dan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Rong Guo
- West China College of Basic Medical Sciences and Forensic Science, Sichuan University, Chengdu, 610041, P. R. China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
18
|
Jia Y, Zhu H, Cai X, Sun C, Ye Y, Cai D, Yang S, Cheng J, Gao J, Yang Y, Zeng H, Zou Q, Li J, Sun H, Wang W. Plant-Derived Immunomodulatory Nanoadjuvants for Cancer Vaccines: Current Status and Future Opportunities. Vaccines (Basel) 2025; 13:378. [PMID: 40333256 PMCID: PMC12031155 DOI: 10.3390/vaccines13040378] [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: 02/07/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 05/09/2025] Open
Abstract
Cancer is a major cause of death worldwide, and vaccine administration is an effective way to stimulate immune responses in patients and to achieve preventive and therapeutic effects. Few vaccines have been used in clinical settings because they have poor immunogenicity, and it is difficult to induce a robust immune response in patients. An adjuvant is an important component of a vaccine that can enhance the intensity, speed, and duration of immune responses. The achievements of adjuvants in the production of stable, safe, and immunogenic tumor vaccines have aroused the enthusiasm of researchers. Recent results have suggested that plant-derived adjuvants have unique advantages, such as greatly improving immune responses to cancer vaccines and promoting humoral and cellular immunity with good biocompatibility and biodegradability. When these adjuvants are used in combination with vaccines, they can not only activate the immune response in vivo but can also promote cytokine secretion and accelerate dendritic cell maturation. This review focused on the application progress of plant adjuvants, including saponins, polysaccharides, flavonoids, and plant virus-like particles, and their combination with nano-delivery systems in cancer vaccines. At the same time, we have also discussed the immunomodulatory mechanisms of these adjuvants and their prospects for improving vaccine efficacy in the treatment of cancer in the future. These promising plant adjuvants may provide prospects and a research basis for the development of tumor vaccines.
Collapse
Affiliation(s)
- Yimin Jia
- Chongqing University Cancer Hospital, Chongqing 400030, China;
| | - Hui Zhu
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Xinyu Cai
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Cun Sun
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Yan Ye
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Dingyi Cai
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
- Department of Stomatology, The 79th Group Army Hospital of PLA, Liaoyang 111000, China
| | - Shuaifei Yang
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Jingjing Cheng
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Jining Gao
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Yun Yang
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Hao Zeng
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Quanming Zou
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Jieping Li
- Affiliated Nanhua Hospital of University of South China, Hengyang 421002, China;
| | - Hongwu Sun
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; (H.Z.); (X.C.); (C.S.); (Y.Y.); (D.C.); (S.Y.); (J.C.); (J.G.); (Y.Y.); (H.Z.); (Q.Z.)
| | - Wenxiu Wang
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou 256600, China
- Shandong Academician Workstation, Binzhou 256600, China
| |
Collapse
|
19
|
Feng L, Zhang J, Ma C, Li K, Zhai J, Cai S, Yin J. Application prospect of polysaccharide in the development of vaccine adjuvants. Int J Biol Macromol 2025; 297:139845. [PMID: 39824409 DOI: 10.1016/j.ijbiomac.2025.139845] [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: 11/24/2024] [Revised: 12/26/2024] [Accepted: 01/12/2025] [Indexed: 01/20/2025]
Abstract
Vaccination is an effective strategy for preventing infectious diseases. Subunit vaccines offer more precise targeting and safer protection compared with traditional inactivated virus vaccines. However, due to their poor immunogenicity, subunit vaccines necessitate the use of adjuvants to stimulate the immune system. Adjuvants have long been incorporated into vaccines to enhance the body's immune response, allowing for reduced dosage and lower production costs. Despite the development of numerous vaccine adjuvants, few exhibit the necessary potency and low toxicity for clinical use, often due to limited efficacy or adverse side effects. This underscores the urgent need for novel human vaccine adjuvants that are safe, effective, and cost-efficient. Recent studies have identified certain natural polysaccharides as promising human vaccine adjuvants due to their immunostimulatory properties, low toxicity, and high safety profiles, which enhance both humoral and cellular immunity. These natural polysaccharides are primarily derived from traditional Chinese medicine (TCM) plants, bacteria, and yeast. This review comprehensively analyzes several promising polysaccharide adjuvants, discussing their clinical applications, market potential, and immunoregulatory activities. In summary, the future prospects of polysaccharides provide valuable insights for the application and development of vaccine adjuvants.
Collapse
Affiliation(s)
- Lei Feng
- Department of Pharmacy, the First Hospital of China Medical University, Shenyang 110001, China; School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Jiarui Zhang
- Department of Intensive Care Medicine, the First Hospital of China Medical University, Shenyang 110001, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, the First Hospital of China Medical University, Shenyang 110001, China
| | - Kai Li
- Department of Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Jianxiu Zhai
- Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Shuang Cai
- Department of Pharmacy, the First Hospital of China Medical University, Shenyang 110001, China; School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Jun Yin
- Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
20
|
Kim EH, Wahl K, Guelfi E, Lee D. Engineering the physical characteristics of biomaterials for innate immune-mediated cancer immunotherapy. J Control Release 2025; 378:814-830. [PMID: 39719214 DOI: 10.1016/j.jconrel.2024.12.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
It has recently been recognized that the physical characteristics of biomaterials - such as size, structure, shape, charge, mechanical strength, hydrophobicity, and multivalency - regulate immunological functions in innate immune cells. In immuno-oncology applications, biomaterials are engineered with distinct physical properties to achieve desired innate immune responses. In this review, we discuss how physical characteristics influence effector functions and innate immune signaling pathways in distinct innate immune cell subtypes. We highlight how physical properties of biomaterials impact phagocytosis regulation, biodistribution, and innate immune cell targeting. We outline the recent advances in physical engineering of biomaterials that directly or indirectly induce desired innate immune responses for cancer immunotherapy. Lastly, we discuss the challenges in current biomaterial approaches that need to be addressed to improve clinical applicability.
Collapse
Affiliation(s)
- Eun-Hye Kim
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Katelyn Wahl
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Erica Guelfi
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - DaeYong Lee
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
21
|
Jia W, Wu Y, Xie Y, Yu M, Chen Y. Advanced Polymeric Nanoparticles for Cancer Immunotherapy: Materials Engineering, Immunotherapeutic Mechanism and Clinical Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413603. [PMID: 39797474 DOI: 10.1002/adma.202413603] [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: 09/10/2024] [Revised: 12/13/2024] [Indexed: 01/13/2025]
Abstract
Cancer immunotherapy, which leverages immune system components to treat malignancies, has emerged as a cornerstone of contemporary therapeutic strategies. Yet, critical concerns about the efficacy and safety of cancer immunotherapies remain formidable. Nanotechnology, especially polymeric nanoparticles (PNPs), offers unparalleled flexibility in manipulation-from the chemical composition and physical properties to the precision control of nanoassemblies. PNPs provide an optimal platform to amplify the potency and minimize systematic toxicity in a broad spectrum of immunotherapeutic modalities. In this comprehensive review, the basics of polymer chemistry, and state-of-the-art designs of PNPs from a physicochemical standpoint for cancer immunotherapy, encompassing therapeutic cancer vaccines, in situ vaccination, adoptive T-cell therapies, tumor-infiltrating immune cell-targeted therapies, therapeutic antibodies, and cytokine therapies are delineated. Each immunotherapy necessitates distinctively tailored design strategies in polymeric nanoplatforms. The extensive applications of PNPs, and investigation of their mechanisms of action for enhanced efficacy are particularly focused on. The safety profiles of PNPs and clinical research progress are discussed. Additionally, forthcoming developments and emergent trends of polymeric nano-immunotherapeutics poised to transform cancer treatment paradigms into clinics are explored.
Collapse
Affiliation(s)
- Wencong Jia
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Ye Wu
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Shanghai Institute of Materdicine, Shanghai, 200051, China
| |
Collapse
|
22
|
Sk MS, Mwangomo R, Daniel L, Gilmore J. Solution Blow Spinning: An Emerging Nanomaterials-Based Wound-Care Technology. J Biomed Mater Res B Appl Biomater 2025; 113:e35513. [PMID: 39854136 DOI: 10.1002/jbm.b.35513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/09/2024] [Accepted: 11/10/2024] [Indexed: 01/26/2025]
Abstract
Application of one-dimensional nanofibers have witnessed exponential growth over the past few decades and are still emerging with their excellent physicochemical and electrical properties. The driving force behind this intriguing transition lies in their unique high surface-to-volume ratio, ubiquitous nanodomains, improved tensile strength, and flexibility to incorporate deliberate functionalities required for specific and advanced applications. Besides numerous benefits, nanomaterials may adversely interact with biological tissues and potentially be cytotoxic and carcinogenic. However, precisely engineered design can outperform the risk with myriad benefits. Wound care technologies are evolving, and products involved in wound care management have a yearly market value of $15-22 billion. Solution blow spinning (SBS) is a facile technique to synthesize biocompatible nanofibers with scalable processing variables for multidirectional biomedical applications. SBS is feasible for a wide range of thermoplastic polymers and nanomaterials to fabricate nanocomposites. This review will focus on the relevance of SBS technology for wound care, including dressings, drug delivery, tissue engineering scaffolds, and sensors.
Collapse
Affiliation(s)
- Md Salauddin Sk
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Ruth Mwangomo
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Luke Daniel
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Jordon Gilmore
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
23
|
Zhao ML, Lei YM, Tang JY, Li W, Cao XY, Liang WB, Yuan R, Yang C, Zhuo Y. DNA lesion-gated dumbbell nanodevices enable on-demand activation of the cGAS-STING pathway for enhancing cancer immunotherapy. Chem Sci 2025; 16:1783-1790. [PMID: 39720145 PMCID: PMC11664422 DOI: 10.1039/d4sc06493c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Utilizing the cGAS-STING pathway to combat immune evasion is one of the most promising strategies for enhancing cancer immunotherapy. However, current techniques for activating the cGAS-STING pathway often face a dilemma, mainly due to the balance between efficacy and safety. Here, we develop a uracil base lesion-gated dumbbell DNA nanodevice (UBLE) that allows on-demand activation and termination of the cGAS-STING pathway in tumor cells, thereby enhancing cancer immunotherapy. The UBLE integrates two deoxyuridines (dU) in the stem for DNA lesion recognition, two locked complementary primer sequences (primers A and B) for DNA self-assembly, and a Förster resonance energy transfer pair (Cy3 and Cy5) attached to the loop for activation assessment. Upon the orthogonal recognition of tumor-specific repair indicators (UDG and APE1), the UBLE undergoes a conformational change to create massive nicked double-stranded DNA (dsDNA) units. These units self-assemble to generate long fluorescent dsDNA structures, permitting selective evaluation and on-demand activation of the cGAS-STING pathway. Furthermore, we demonstrate that the UBLE can effectively activate the cGAS-STING pathway in tumor cells, enhancing NK cell-targeted cancer immunotherapy. This work develops a DNA lesion-gated strategy for on-demand activation and termination of the cGAS-STING pathway, affording an innovative avenue for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Mei-Ling Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Yan-Mei Lei
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 2002127 P. R. China
| | - Jing-Yi Tang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Wen Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Xin-Yu Cao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Wen-Bin Liang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Chaoyong Yang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 2002127 P. R. China
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 P. R. China
| | - Ying Zhuo
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| |
Collapse
|
24
|
Pereira DR, Pérez-Betancourt Y, Távora BCLF, Magalhães GS, Carmona-Ribeiro AM, Faquim-Mauro EL. The Role of Dendritic Cells in Adaptive Immune Response Induced by OVA/PDDA Nanoparticles. Vaccines (Basel) 2025; 13:76. [PMID: 39852855 PMCID: PMC11769024 DOI: 10.3390/vaccines13010076] [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: 10/31/2024] [Revised: 12/08/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
Background/Objective: Cationic polymers were shown to assemble with negatively charged proteins yielding nanoparticles (NPs). Poly-diallyl-dimethyl-ammonium chloride (PDDA) combined with ovalbumin (OVA) yielded a stable colloidal dispersion (OVA/PDDA-NPs) eliciting significant anti-OVA immune response. Dendritic cells (DCs), as sentinels of foreign antigens, exert a crucial role in the antigen-specific immune response. Here, we aimed to evaluate the involvement of DCs in the immune response induced by OVA/PDDA. Methods: In vivo experiments were used to assess the ability of OVA/PDDA-NPs to induce anti-OVA antibodies by ELISA, as well as plasma cells and memory B cells using flow cytometry. Additionally, DC migration to draining lymph nodes following OVA/PDDA-NP immunization was evaluated by flow cytometry. In vitro experiments using bone marrow-derived DCs (BM-DCs) were used to analyze the binding and uptake of OVA/PDDA-NPs, DC maturation status, and their antigen-presenting capacity. Results: Our data confirmed the potent effect of OVA/PDDA-NPs inducing anti-OVA IgG1 and IgG2a antibodies with increased CD19+CD138+ plasma cells and CD19+CD38+CD27+ memory cells in immunized mice. OVA/PDDA-NPs induced DC maturation and migration to draining lymph nodes. The in vitro results showed higher binding and the uptake of OVA/PDDA-NPs by BM-DCs. In addition, the NPs were able to induce the upregulation of costimulatory and MHC-II molecules on DCs, as well as TNF-α and IL-12 production. Higher OVA-specific T cell proliferation was promoted by BM-DCs incubated with OVA/PDDA-NPs. Conclusions: The data showed the central role of DCs in the induction of antigen-specific immune response by OVA-PDDA-NPs, thus proving that these NPs are a potent adjuvant for subunit vaccine design.
Collapse
Affiliation(s)
- Daniele R. Pereira
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05585-000, Brazil; (D.R.P.); (B.C.L.F.T.); (G.S.M.)
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 04021-001, Brazil
| | - Yunys Pérez-Betancourt
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo 04021-001, Brazil; (Y.P.-B.); (A.M.C.-R.)
| | - Bianca C. L. F. Távora
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05585-000, Brazil; (D.R.P.); (B.C.L.F.T.); (G.S.M.)
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 04021-001, Brazil
| | - Geraldo S. Magalhães
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05585-000, Brazil; (D.R.P.); (B.C.L.F.T.); (G.S.M.)
| | - Ana Maria Carmona-Ribeiro
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo 04021-001, Brazil; (Y.P.-B.); (A.M.C.-R.)
| | - Eliana L. Faquim-Mauro
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05585-000, Brazil; (D.R.P.); (B.C.L.F.T.); (G.S.M.)
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 04021-001, Brazil
| |
Collapse
|
25
|
Shariati A, Khezrpour A, Shariati F, Afkhami H, Yarahmadi A, Alavimanesh S, Kamrani S, Modarressi MH, Khani P. DNA vaccines as promising immuno-therapeutics against cancer: a new insight. Front Immunol 2025; 15:1498431. [PMID: 39872522 PMCID: PMC11769820 DOI: 10.3389/fimmu.2024.1498431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Cancer is one of the leading causes of mortality around the world and most of our conventional treatments are not efficient enough to combat this deadly disease. Harnessing the power of the immune system to target cancer cells is one of the most appealing methods for cancer therapy. Nucleotide-based cancer vaccines, especially deoxyribonucleic acid (DNA) cancer vaccines are viable novel cancer treatments that have recently garnered significant attention. DNA cancer vaccines are made of plasmid molecules that encode tumor-associated or tumor-specific antigens (TAAs or TSAs), and possibly some other immunomodulatory adjuvants such as pro-inflammatory interleukins. Following the internalization of plasmids into cells, their genes are expressed and the tumor antigens are loaded on major histocompatibility molecules to be presented to T-cells. After the T-cells have been activated, they will look for tumor antigens and destroy the tumor cells upon encountering them. As with any other treatment, there are pros and cons associated with using these vaccines. They are relatively safe, usually well-tolerated, stable, easily mass-produced, cost-effective, and easily stored and transported. They can induce a systemic immune response effective on both the primary tumor and metastases. The main disadvantage of DNA vaccines is their poor immunogenicity. Several approaches including structural modification, combination therapy with conventional and novel cancer treatments (such as chemotherapy, radiotherapy, and immune checkpoint blockade (ICB)), and the incorporation of adjuvants into the plasmid structure have been studied to enhance the vaccine's immunogenicity and improve the clinical outcome of cancer patients. In this review, we will discuss some of the most promising optimization strategies and examine some of the important trials regarding these vaccines.
Collapse
Affiliation(s)
- Alireza Shariati
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Arya Khezrpour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Fatemeh Shariati
- Department of Genetics, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
26
|
Ghattas M, Dwivedi G, Chevrier A, Horn-Bourque D, Alameh MG, Lavertu M. Chitosan immunomodulation: insights into mechanisms of action on immune cells and signaling pathways. RSC Adv 2025; 15:896-909. [PMID: 39802469 PMCID: PMC11719903 DOI: 10.1039/d4ra08406c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025] Open
Abstract
Chitosan, a biodegradable and biocompatible natural polymer composed of β-(1-4)-linked N-acetyl glucosamine (GlcNAc) and d-glucosamine (GlcN) and derived from crustacean shells, has been widely studied for various biomedical applications, including drug delivery, cartilage repair, wound healing, and tissue engineering, because of its unique physicochemical properties. One of the most promising areas of research is the investigation of the immunomodulatory properties of chitosan, since the biopolymer has been shown to modulate the maturation, activation, cytokine production, and polarization of dendritic cells and macrophages, two key immune cells involved in the initiation and regulation of innate and adaptive immune responses, leading to enhanced immune responses. Several signaling pathways, including the cGAS-STING, STAT-1, and NLRP3 inflammasomes, are involved in chitosan-induced immunomodulation. This review provides a comprehensive overview of the current understanding of the in vitro immunomodulatory effects of chitosan. This information may facilitate the development of chitosan-based therapies and vaccine adjuvants for various immune-related diseases.
Collapse
Affiliation(s)
- Majed Ghattas
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Garima Dwivedi
- Perelman School of Medicine, University of Pennsylvania Philadelphia PA USA
| | - Anik Chevrier
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Delano Horn-Bourque
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Mohamad-Gabriel Alameh
- Perelman School of Medicine, University of Pennsylvania Philadelphia PA USA
- Penn Institute for RNA Innovation, University of Pennsylvania Philadelphia PA USA
| | - Marc Lavertu
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| |
Collapse
|
27
|
Owliaee I, Khaledian M, Shojaeian A, Madanchi H, Yarani R, Boroujeni AK, Shoushtari M. Antimicrobial Peptides Against Arboviruses: Mechanisms, Challenges, and Future Directions. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10430-0. [PMID: 39776036 DOI: 10.1007/s12602-024-10430-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
This review delves into the potential of antimicrobial peptides (AMPs) as promising candidates for combating arboviruses, focusing on their mechanisms of antiviral activity, challenges, and future directions. AMPs have shown promise in preventing arbovirus attachment to host cells, inducing interferon production, and targeting multiple viral stages, illustrating their multifaceted impact on arbovirus infections. Structural elucidation of AMP-viral complexes is explored to deepen the understanding of molecular determinants governing viral neutralization, paving the way for structure-guided design. Furthermore, this review highlights the potential of AMP-based combination therapies to create synergistic effects that enhance overall treatment outcomes while minimizing the likelihood of resistance development. Challenges such as susceptibility to proteases, toxicity, and scalable production are discussed alongside strategies to address these limitations. Additionally, the expanding applications of AMPs as vaccine adjuvants and antiviral delivery systems are emphasized, underscoring their versatility beyond direct antiviral functions.
Collapse
Affiliation(s)
- Iman Owliaee
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, 65178-38736, Iran
- Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, 65178-38736, Iran
| | - Mehran Khaledian
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, 65178-38736, Iran
- Department of Medical Entomology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, 65178-38736, Iran
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Hamid Madanchi
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, 35147-99442, Iran
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 13169-43551, Iran
| | - Reza Yarani
- Interventional Radiology Innovation at Stanford (IRIS), Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Armin Khaghani Boroujeni
- Skin Disease and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Shoushtari
- Department of Virology, Pasteur Institute of Iran, Tehran, 13169-43551, Iran.
| |
Collapse
|
28
|
Lian Z, Liu X, Li X. Elucidating the expression and role of cGAS in pan-cancer using integrated bioinformatics and experimental approaches. BMC Cancer 2025; 25:5. [PMID: 39748320 PMCID: PMC11697830 DOI: 10.1186/s12885-024-13379-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: 05/28/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
cGAS plays an important role in regulating both tumor immune responses and DNA damage repair. Nevertheless, there was little research that comprehensively analyzed the correlation between cGAS and the tumor microenvironment, immune cell infiltration, and DNA damage repair in different cancers. In this study, The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) data were used to analyze the mRNA expression and genomic alterations of cGAS in pan-cancer. The HPA database was used to explore the protein levels of cGAS in normal tissues and cancers. Correlation analysis were performed to explore the role of cGAS in interferon expression, immune cell infiltrations, DNA damage repair, and predictive immune markers. The prognostic value of cGAS was analyzed using survival data from the TCGA, Kaplan-Meier plotter database, and PrognoScan database. Lastly, the role of cGAS in DNA damage repair signaling and interferon signaling was validated in NSCLC cell lines. The results showed that cGAS was widely expressed in human normal tissues and various cancers, and the expression of cGAS was significantly upregulated in almost all of the solid cancers. Genomic analysis indicated that the expression of cGAS was positively correlated with copy number levels, while negatively correlated with the methylation levels of cGAS promoter. In addition, the level of cGAS was positively correlated with type I interferons expression, infiltration levels of most immune cell types, TMB and MSI levels, stromal and immune scores, and DNA damage repair gene sets including nonhomologous end joining and homologous recombination pathway. Survival analysis indicated that cGAS levels were associated with patient prognosis in several cancers. Lastly, in vitro study showed knockdown of cGAS expression inhibits the DNA damage repair signaling pathway and interferon signaling in NSCLC. In conclusions, cGAS is wildly activated in human cancers, which might participate in regulating cancer immunity and DNA damage repair. cGAS could be used as an effective target for cancer treatment and might be a potential predictive immune marker.
Collapse
Affiliation(s)
- Zhen Lian
- Department of Emergency, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Xue Liu
- Department of Comprehensive Treatment Ward, Mudan People's Hospital of Heze, Heze, 274000, China
| | - Xue Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| |
Collapse
|
29
|
Liang J, Yao L, Liu Z, Chen Y, Lin Y, Tian T. Nanoparticles in Subunit Vaccines: Immunological Foundations, Categories, and Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407649. [PMID: 39501996 DOI: 10.1002/smll.202407649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/12/2024] [Indexed: 01/11/2025]
Abstract
Subunit vaccines, significant in next-generation vaccine development, offer precise targeting of immune responses by focusing on specific antigens. However, this precision often comes at the cost of eliciting strong and durable immunity, posing a great challenge to vaccine design. To address this limitation, recent advancements in nanoparticles (NPs) are utilized to enhance antigen delivery efficiency and boost vaccine efficacy. This review examines how the physicochemical properties of NPs influence various stages of the immune response during vaccine delivery and analyzes how different NP types contribute to immune activation and enhance vaccine performance. It then explores the unique characteristics and immune activation mechanisms of these NPs, along with their recent advancements, and highlights their application in subunit vaccines targeting infectious diseases and cancer. Finally, it discusses the challenges in NP-based vaccine development and proposes future directions for innovation in this promising field.
Collapse
Affiliation(s)
- Jiale Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lan Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ye Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West ChinaHospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Taoran Tian
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan, 610041, China
| |
Collapse
|
30
|
Wang C, Zhang X, Li Q, Hou Y, Sun M, Sun J, Lou Z, Han X, Li Y. A review of carbohydrate polymer-synthesized nanoparticles in cancer immunotherapy: Past, present and future perspectives. Int J Biol Macromol 2025; 286:138195. [PMID: 39645110 DOI: 10.1016/j.ijbiomac.2024.138195] [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: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Cancer continues to be a leading factor in mortality and tackling it has been made difficult by the development of immune escape. Furthermore, alternative treatments like surgery, chemotherapy, and radiation have been unsuccessful in eradicating cancer. Despite being effective, they have not succeeded in providing a full cancer treatment and exhibit several negative effects. The field of immunotherapy has been improved by utilizing cancer vaccines, immune checkpoint inhibitors (ICIs), and adoptive cell transfer to enhance immune responses to tumors. Nevertheless, cancer cells need to adapt and become immune to immune reactions, leading to the need for innovative treatment methods. Carbohydrate polymers and their nanoparticles have been beneficial in improving cancer immunotherapy by being customizable to specifically target the immune system. These nanoparticles can change the tumor microenvironment and accelerate immunotherapy by affecting immune cells such as T cells and dendritic cells. Incorporating both chemotherapy and phototherapy into nanoparticles can improve immunotherapy. Furthermore, besides controlling immune reactions, carbohydrate polymer nanoparticles can also be used for theranostic purposes, where they are used to image tumor cells and activate the immune system to eradicate cancer.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China
| | - Xueyao Zhang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China
| | - Qiaobei Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China
| | - Yuxin Hou
- Department of Ultrasonic Diagnosis, The Benxi Hospital of China Medical University, Benxi, China
| | - Minglu Sun
- Department of Ultrasonic Diagnosis, The Cancer Hospital of China Medical University, Shenyang, China
| | - Jun Sun
- Department of Intervention, the Fourth Hospital of China Medical University, Shenyang, China
| | - Zhe Lou
- Department of Cardiovascular Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
31
|
Elkhenany H, Soliman MW, Atta D, El-Badri N. Innovative Marine-Sourced Hydroxyapatite, Chitosan, Collagen, and Gelatin for Eco-Friendly Bone and Cartilage Regeneration. J Biomed Mater Res A 2025; 113:e37833. [PMID: 39508545 DOI: 10.1002/jbm.a.37833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024]
Abstract
In recent years, the exploration of sustainable alternatives in the field of bone tissue engineering has led researchers to focus on marine waste byproducts as a valuable resource. These marine resources, often overlooked remnants of various industries, exhibit a rich composition of hydroxyapatite, collagen, calcium carbonate, and other minerals essential to the complex framework of bone structure. Marine waste by-products can emit gases such as methane and carbon dioxide, highlighting the urgency to repurpose these materials for innovative tissue regeneration solutions, offering a sustainable approach to address environmental challenges while advancing medical science. Using these discarded materials offers a promising pathway for sustainable development in regenerative medicine. This review investigates the distinctive properties of marine waste byproducts, emphasizing their capacity to be recycled effectively to contribute to the rebuilding of bone and cartilage tissue during regeneration processes. We also highlight the compatibility of these resources with biological materials such as platelet-rich plasma (PRP), stem cells, exosomes, and natural bioproducts, as well as nanoparticles (NPs) and polymers. By using the natural potential of these resources, we simultaneously address environmental challenges and promote innovative solutions in skeletal tissue engineering, initiating a new era of environmentally green biomedical research.
Collapse
Affiliation(s)
- Hoda Elkhenany
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Mariam Waleed Soliman
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| | - Dina Atta
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
32
|
Xu S, Zhao Z, Sun C, Ji Y, Luan Q, Zhang Q, Jin Z, Zhao K. Immunoprotective effect of chitosan nanoparticles with different particle sizes against H9N2 avian influenza infection. Poult Sci 2025; 104:104559. [PMID: 39603189 PMCID: PMC11635735 DOI: 10.1016/j.psj.2024.104559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
H9N2 is the most common avian influenza virus (AIV), which causes significant losses in chickens. Safe and effective vaccines are crucial for the prevention of H9N2 AIVs. Chitosan nanoparticles, as novel adjuvants, enhance vaccine immunity and biocompatibility; however, the impact of particle size on the immunological effects remains underexplored. To solve these problems and to prepare an efficient novel H9N2 vaccine, we constructed four N-2-HACC/CMCS NPs (NHC NPs) of different particle sizes (165.6 ± 12.0 nm, 272.5 ± 7.0 nm, 343.2 ± 8.0 nm, and 443.5 ± 15.0 nm). Subsequent in vivo immunogenicity screening revealed that H9N2 with the 272.5 ± 7.0 nm NHC NPs vaccine group induced higher levels of neutralizing antibodies in the early stage of the immune response, while the 343.2 ± 8.0 nm NHC NPs vaccine group induced higher levels of neutralizing antibodies in the late stages of the immune response. Subsequently, the results of the optimal particle size combination screening revealed that more neutralizing antibodies were induced when the NHC NPs particle size combination of 272.5 ± 7.0 nm:343.2 ± 8.0 nm ratio was 1.5:1. This optimal particle size combination for NP vaccines promoted lymphocyte proliferation, induced higher IgG2a/IgG1 ratios, and promoted the production of cytokines (i.e., IL-2, IL-4, and IFN-γ). Moreover, a mechanistic analysis revealed that the optimal NHC NPs combination triggered the activation of antigen presenting cells via TLR4 and participated in immune responses through the production of NO and TNF-α. Taken together, our study revealed that the optimal combination of NHC NPs may be a promising strategy against influenza viruses.
Collapse
Affiliation(s)
- Shangen Xu
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Zhi Zhao
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Chenxi Sun
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Yile Ji
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Qingshuang Luan
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Qihong Zhang
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Zheng Jin
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Kai Zhao
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China.
| |
Collapse
|
33
|
Reay SL, Marina Ferreira A, Hilkens CMU, Novakovic K. The Paradoxical Immunomodulatory Effects of Chitosan in Biomedicine. Polymers (Basel) 2024; 17:19. [PMID: 39795422 PMCID: PMC11723117 DOI: 10.3390/polym17010019] [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: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Chitosan is widely explored in the field of biomedicine due to its abundance and reported properties, including biocompatibility, biodegradability, non-toxicity, mucoadhesion, and anti-microbial activity. Although our understanding of the immune response to chitosan has evolved, confusion remains regarding whether chitosan is a pro- or anti-inflammatory biomaterial. Tackling this knowledge gap is essential for the translation of chitosan-based biomaterials to clinical use. Herein, we provide an overview of the immune responses to chitosan, exploring the roles of endotoxin contamination and physiochemical properties in immunomodulation. Ultimately, this literature review concludes that various physiochemical properties, including molecular weight, degree of deacetylation and polydispersity, endotoxin contamination, and cellular environment, interplay in the complex process of chitosan immunomodulation, which can lead to both pro- and anti-inflammatory effects.
Collapse
Affiliation(s)
- Sophie L. Reay
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| | - Catharien M. U. Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Katarina Novakovic
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| |
Collapse
|
34
|
Hoover AR, Liu K, Furrer C, Lam SSK, Anderson DW, Zhou Z, Yang J, Wong CF, Medcalf AD, Sun XH, Hode T, Alleruzzo L, Delawder A, Raker J, Abousleiman G, Valerio TI, Sun Y, Papin JF, Li M, Chen WR. N-Dihydrogalactochitosan Drives Conventional and Alternative Activations of STING to Synergize Type I IFN and IL-1β Productions for Antitumor Immunity. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2410079. [PMID: 39896882 PMCID: PMC11784597 DOI: 10.1002/adfm.202410079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Indexed: 02/04/2025]
Abstract
N-dihydrogalactochitosan (GC) is an immune stimulant/adjuvant. Synthesized from chitosan and galactose, GC is a new chemical entity that significantly enhances the immune-stimulating properties of its parental material, chitosan, making it a promising therapeutic agent. When used in combination with antigenic material, GC stimulates innate and adaptive antitumor and antiviral immunities. However, its mechanism has not been fully investigated. Herein we demonstrate that GC drives type I IFN activation in antigen-presenting cells (APCs). More importantly, GC drives alternative STING pathways, leading to inflammatory cell death that enhances dendritic cell (DC) activation. GC-activated DCs trigger a variety of nucleic acid sensing pattern recognition receptors (PRRs) pathways and IL-1β production via the activation of the inflammasome. In vivo, GC induces a potent response of type I IFNs and upregulates genes associated with STING signaling within the tumor microenvironment (TME). Moreover, intratumoral delivery of GC reduces the numbers of M2-like macrophages and increases M1-like macrophages residing within the TME, while subsequently increasing the number of activated DCs. Our findings demonstrate that GC acts as a multimodal immune stimulant via STING to generate a broad type I IFN response. This uniquely broad response holds therapeutic promise in generating enhanced antitumor and antiviral immunities.
Collapse
Affiliation(s)
- Ashley R. Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | | | | | - Zhijun Zhou
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | - Alexandra D. Medcalf
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xiao-Hong Sun
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Tomas Hode
- Immunophotonics, Inc., Saint Louis, MO, USA
| | | | | | | | - Ghainaa Abousleiman
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I. Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Yuanhong Sun
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - James F. Papin
- Department Pathology and Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Wei R. Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
35
|
Farag EAH, Baromh MZ, El-Kalamwi N, Sherif AH. Vitamin E nanoparticles enhance performance and immune status of Nile tilapia. BMC Vet Res 2024; 20:561. [PMID: 39668352 PMCID: PMC11636036 DOI: 10.1186/s12917-024-04398-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] [Received: 09/06/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Vitamin E (VE) is an essential vitamin liposoluble antioxidant in aquatic animals that is usually lost during feed processing and digestion, whereas nano-chitosan, a polysaccharide, could protect VE. In this study, Nile tilapia (70.85 ± 0.2 g) was fed VE (100 mg/kg dry diet) and a chitosan protected-VE nanoparticle (NPs) with gradual percentages of recommended dose 25%, 50%, 75%, and 100% for 4, 6, and 8 weeks. Growth parameters total weight gain (TG), daily weight gain (DWG), and relative growth rate (RGR) were significantly and positively correlated with VENPs additions. Regardless of the addition level, the feed conversion ratio (FCR) was significantly lower in the VENP groups. Lysozyme, serum antibacterial activity, and oxidative burst activity indicated the superiority of VENPs (VENPs75 and VENPs100) in enhancing the fish's innate immunity compared to bulk VE and the control groups. Fish were experimentally challenged with pathogenic Aeromonas hydrophila; those received dietary showed a low mortality rate (MR%), about 40% compared with 70% in the control with lower re-isolation compared to the control and VE groups. VENPs could provide ascending relative protection level during the period of 4 to 8 weeks; RPL ranged from 33.3 to 42.86% (VENPs100), 16.67-42.86% (VENPs75), 0 to 28.57% (VENPs50), and 0 to 14.29% (VENPs25 and VE), respectively. Finally, this study recommended incorporating VENPs into the Nile tilapia diet at 50, 75, and 100 mg/ kg fish feed. Fish in the VENPs75 and VENPs100 groups were immune boosted, becoming less vulnerable to A. hydrophila infection.
Collapse
Affiliation(s)
- Enas A H Farag
- Department of Pharmacology, Animal Health Research Institute AHRI, Agriculture Research Center ARC, Benha, Egypt
| | - Mohamed Z Baromh
- Division of Aquaculture, National Institute of Oceanography and Fisheries (NIOF), Alexandria, Egypt
| | - Naglaa El-Kalamwi
- Pathology Department, Animal Health Research Institute AHRI, Agriculture Research Center ARC, Dokki, Egypt
| | - Ahmed H Sherif
- Fish Diseases Department, Animal Health Research Institute AHRI, Agriculture Research Centre ARC, Kafrelsheikh, Egypt.
| |
Collapse
|
36
|
Liang X, Zhou J, Wang M, Wang J, Song H, Xu Y, Li Y. Progress and prospect of polysaccharides as adjuvants in vaccine development. Virulence 2024; 15:2435373. [PMID: 39601191 PMCID: PMC11622597 DOI: 10.1080/21505594.2024.2435373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024] Open
Abstract
Vaccines are an effective approach to confer immunity against infectious diseases. Modern subunit vaccines offer more precise target and safe protection compared to traditional whole-pathogen vaccines. However, subunit vaccines require adjuvants to stimulate the immune system due to the less immunogenicity. Adjuvants strengthen immunogenicity by enhancing, modulating, and prolonging the immune response. Unfortunately, few adjuvants have sufficient potency and low enough toxicity for clinical use, highlighting the urgent need for new vaccine adjuvants with the characteristics of safety, efficacy, and cost-effectiveness. Notably, some natural polysaccharides have been approved as adjuvants in human vaccines, owing to their intrinsic immunomodulation, low toxicity, and high safety. Natural polysaccharides are mainly derived from plants, bacteria, and yeast. Partly owing to the difficulty of obtaining them, synthetic polysaccharides emerged in clinical trials. The immune mechanisms of both natural and synthetic polysaccharides remain incompletely understood, hindering the rational development of polysaccharide adjuvants. This comprehensive review primarily focused on several promising polysaccharide adjuvants, discussing their recent applications in vaccines and highlighting their immune-modulatory effects. Furthermore, the future perspectives of polysaccharides offer insightful guidance to adjuvant development and application.
Collapse
Affiliation(s)
- Xinlong Liang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Jiaying Zhou
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Mengmeng Wang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Jing Wang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Houhui Song
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Yigang Xu
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
| | - Yuan Li
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, China
- Research and Development Department, Zhejiang Huijia Biotechnology Co. Ltd ., Huzhou, People’s Republic of China
| |
Collapse
|
37
|
Wu L, Xu W, Jiang H, Yang M, Cun D. Respiratory delivered vaccines: Current status and perspectives in rational formulation design. Acta Pharm Sin B 2024; 14:5132-5160. [PMID: 39807330 PMCID: PMC11725141 DOI: 10.1016/j.apsb.2024.08.026] [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: 05/25/2024] [Revised: 07/20/2024] [Accepted: 08/18/2024] [Indexed: 01/16/2025] Open
Abstract
The respiratory tract is susceptible to various infections and can be affected by many serious diseases. Vaccination is one of the most promising ways that prevent infectious diseases and treatment of some diseases such as malignancy. Direct delivery of vaccines to the respiratory tract could mimic the natural process of infection and shorten the delivery path, therefore unique mucosal immunity at the first line might be induced and the efficiency of delivery can be high. Despite considerable attempts at the development of respiratory vaccines, the rational formulation design still warrants attention, i.e., how the formulation composition, particle properties, formulation type (liquid or solid), and devices would influence the immune outcome. This article reviews the recent advances in the formulation design and development of respiratory vaccines. The focus is on the state of the art of delivering antigenic compounds through the respiratory tract, overcoming the pulmonary bio-barriers, enhancing delivery efficiencies of respiratory vaccines as well as maintaining the stability of vaccines during storage and use. The choice of devices and the influence of deposition sites on vaccine efficiencies were also reviewed.
Collapse
Affiliation(s)
- Lan Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Wenwen Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Huiyang Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang 110016, China
- School of Food and Drug, Shenzhen Polytechnic University, China, Shenzhen 518055, China
| |
Collapse
|
38
|
Thu VTA, Hoang TX, Park JK, Kim JY. Induction of Innate Immune Memory in LPS-Primed Microglial Cells by Water-Soluble Chitosan. BIOMED RESEARCH INTERNATIONAL 2024; 2024:8027006. [PMID: 39654846 PMCID: PMC11628173 DOI: 10.1155/bmri/8027006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/24/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Innate immune memory or trained immunity refers to a long-lasting response of the innate immune cells against repeated exposure to the homogenous or heterogenous infectious agent. The trained immunity is induced through epigenetic modification and is characterized by the change of both intracellular immunological signaling and cellular metabolism. Recently, different groups have tried to establish protocols to generate trained innate immune cells. However, the molecular basis of innate memory induction remains poorly understood. Here, we evaluated the impact of water-soluble chitosan on the innate immune memory induction in microglial cells primed with LPS. The trained-immune response was accessed by measuring proinflammatory markers, metabolic change, and epigenetic modification. We showed that the stimulation/restimulation with LPS only caused a robust reduction of iNOS, and proinflammatory cytokines, indicating induced immune tolerance. In contrast, the treatment of chitosan induces long-lasting memory microglial cells accompanied by a high level of iNOS, increased lactate production, induced epigenetic modification, and the upregulation of proinflammatory cytokines upon further exposure to the same stimulus. These findings suggest that chitosan induces microglial-trained immunity by targeting distinct epigenetic and metabolic pathways; therefore, chitosan treatment may provide a novel approach for targeting innate immunity towards a memory-like response in an in vitro model.
Collapse
Affiliation(s)
- Vo Thuy Anh Thu
- Department of Life Science, Gachon University, Seongnam, Gyeonggi-do 13120, Republic of Korea
| | - Thi Xoan Hoang
- Department of Life Science, Gachon University, Seongnam, Gyeonggi-do 13120, Republic of Korea
| | - Jae Kweon Park
- Department of Life Science, Gachon University, Seongnam, Gyeonggi-do 13120, Republic of Korea
| | - Jae Young Kim
- Department of Life Science, Gachon University, Seongnam, Gyeonggi-do 13120, Republic of Korea
| |
Collapse
|
39
|
Weth AF, Dangerfield EM, Timmer MSM, Stocker BL. Recent Advances in the Development of Mincle-Targeting Vaccine Adjuvants. Vaccines (Basel) 2024; 12:1320. [PMID: 39771982 PMCID: PMC11680293 DOI: 10.3390/vaccines12121320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 01/11/2025] Open
Abstract
The Macrophage-inducible C-type lectin (Mincle) is a pattern-recognition receptor (PRR), which has shown much promise as a molecular target for the development of TH1/TH17-skewing vaccine adjuvants. In 2009, the first non-proteinaceous Mincle ligands, trehalose dimycolate (TDM) and trehalose dibehenate (TDB), were identified. This prompted a search for other Mincle agonists and the exploration of Mincle agonists as vaccine adjuvants for both preventative and therapeutic (anti-cancer) vaccines. In this review, we discuss those classes of Mincle agonists that have been explored for their adjuvant potential. These Mincle agonists have been used as stand-alone adjuvants or in combination with other pathogen-associated molecular patterns (PAMPs) or immunomodulatory agents. We will also highlight recently identified Mincle ligands with hitherto unknown adjuvanticity. Conjugate vaccines that contain covalently linked adjuvants and/or adjuvant-antigen combinations are also presented, as well as the different formulations (e.g., oil-in-water emulsions, liposomes, and particulate delivery systems) that have been used for the codelivery of antigens and adjuvants. Insofar the reader is presented with a thorough review of the potential of Mincle-mediated vaccine adjuvants, including historical context, present-day research and clinical trials, and outstanding research questions, such as the role of ligand presentation and Mincle clustering, which, if better understood, will aid in the development of the much-needed TH1/TH17-skewing vaccine adjuvants.
Collapse
Affiliation(s)
| | | | - Mattie S. M. Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Bridget L. Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| |
Collapse
|
40
|
Gamberi C, Leverette CL, Davis AC, Ismail M, Piccialli I, Borbone N, Oliviero G, Vicidomini C, Palumbo R, Roviello GN. Oceanic Breakthroughs: Marine-Derived Innovations in Vaccination, Therapy, and Immune Health. Vaccines (Basel) 2024; 12:1263. [PMID: 39591167 PMCID: PMC11598900 DOI: 10.3390/vaccines12111263] [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: 10/10/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The vast, untapped potential of the world's oceans is revealing groundbreaking advancements in human health and vaccination. Microalgae such as Nannochloropsis spp. and Dunaliella salina are emerging as resources for recombinant vaccine development with specific and heterologous genetic tools used to boost production of functional recombinant antigens in Dunaliella salina and Nannochloropsis spp. to induce immunoprotection. In humans, several antigens produced in microalgae have shown potential in combating diseases caused by the human papillomavirus, human immunodeficiency virus, hepatitis B virus, influenza virus, Zika virus, Zaire Ebola virus, Plasmodium falciparum, and Staphylococcus aureus. For animals, microalgae-derived vaccine prototypes have been developed to fight against the foot-and-mouth disease virus, classical swine fever virus, vibriosis, white spot syndrome virus, and Histophilus somni. Marine organisms offer unique advantages, including the ability to express complex antigens and sustainable production. Additionally, the oceans provide an array of bioactive compounds that serve as therapeutics, potent adjuvants, delivery systems, and immunomodulatory agents. These innovations from the sea not only enhance vaccine efficacy but also contribute to broader immunological and general health. This review explores the transformative role of marine-derived substances in modern medicine, emphasizing their importance in the ongoing battle against infectious diseases.
Collapse
Affiliation(s)
- Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Chad L. Leverette
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Alexis C. Davis
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA; (C.G.); (C.L.L.); (A.C.D.)
| | - Moayad Ismail
- Faculty of Medicine, European University, 76 Guramishvili Ave., 0141 Tbilisi, Georgia;
| | - Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Nicola Borbone
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Giorgia Oliviero
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Caterina Vicidomini
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| | - Rosanna Palumbo
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging, Italian National Research Council (IBB-CNR), Via P. Castellino 111, 80131 Naples, Italy; (C.V.); (R.P.)
| |
Collapse
|
41
|
Berkenfeld K, Carneiro S, Corzo C, Laffleur F, Salar-Behzadi S, Winkeljann B, Esfahani G. Formulation strategies, preparation methods, and devices for pulmonary delivery of biologics. Eur J Pharm Biopharm 2024; 204:114530. [PMID: 39393712 DOI: 10.1016/j.ejpb.2024.114530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Biological products, including vaccines, blood components, and recombinant therapeutic proteins, are derived from natural sources such as humans, animals, or microorganisms and are typically produced using advanced biotechnological methods. The success of biologics, particularly monoclonal antibodies, can be attributed to their favorable safety profiles and target specificity. However, their large molecular size presents significant challenges in drug delivery, particularly in overcoming biological barriers. Pulmonary delivery has emerged as a promising route for administering biologics, offering non-invasive delivery with rapid absorption, high systemic bioavailability, and avoidance of first-pass metabolism. This review first details the anatomy and physiological barriers of the respiratory tract and the associated challenges of pulmonary drug delivery (PDD). It further discusses innovations in PDD, the impact of particle size on drug deposition, and the use of secondary particles, such as nanoparticles, to enhance bioavailability and targeting. The review also explains various devices used for PDD, including dry powder inhalers (DPIs) and nebulizers, highlighting their advantages and limitations in delivering biologics. The role of excipients in improving the stability and performance of inhalation products is also addressed. Since dry powders are considered the suitable format for delivering biomolecules, particular emphasis is placed on the excipients used in DPI development. The final section of the article reviews and compares various dry powder manufacturing methods, clarifying their clinical relevance and potential for future applications in the field of inhalable drug formulation.
Collapse
Affiliation(s)
- Kai Berkenfeld
- Laboratory of Pharmaceutical Technology and Biopharmaceutics, Institute of Pharmacy, University of Bonn, Gerhard-Domagk-Street 3, 53121 Bonn, Germany; Pharmaceutical Engineering and Technology Research Scientists (PETRS)
| | - Simone Carneiro
- Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Haus B, 81377 München, Germany; Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, München 80799, Germany; Pharmaceutical Engineering and Technology Research Scientists (PETRS)
| | - Carolina Corzo
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria; Pharmaceutical Engineering and Technology Research Scientists (PETRS)
| | - Flavia Laffleur
- Department of Pharmaceutical Technology, Institute of Pharmacy, Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; Pharmaceutical Engineering and Technology Research Scientists (PETRS)
| | - Sharareh Salar-Behzadi
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria; Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology, University of Graz, Graz, Austria; Pharmaceutical Engineering and Technology Research Scientists (PETRS)
| | - Benjamin Winkeljann
- Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Haus B, 81377 München, Germany; Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, München 80799, Germany; RNhale GmbH, München 81371, Germany; Comprehensive Pneumology Center Munich (CPC-M), Helmholtz Munich, German Center for Lung Research (DZL), 81377 Munich, Germany; Pharmaceutical Engineering and Technology Research Scientists (PETRS)
| | - Golbarg Esfahani
- Department of Pharmaceutical Technology, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Straße 4, Halle 06120, Saale, Germany; Pharmaceutical Engineering and Technology Research Scientists (PETRS).
| |
Collapse
|
42
|
Mu L, Wu L, Wu S, Ye Q, Zhong Z. Progress in chitin/chitosan and their derivatives for biomedical applications: Where we stand. Carbohydr Polym 2024; 343:122233. [PMID: 39174074 DOI: 10.1016/j.carbpol.2024.122233] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/24/2024] [Accepted: 05/02/2024] [Indexed: 08/24/2024]
Abstract
Chitin and its deacetylated form, chitosan, have demonstrated remarkable versatility in the realm of biomaterials. Their exceptional biocompatibility, antibacterial properties, pro- and anticoagulant characteristics, robust antioxidant capacity, and anti-inflammatory potential make them highly sought-after in various applications. This review delves into the mechanisms underlying chitin/chitosan's biological activity and provides a comprehensive overview of their derivatives in fields such as tissue engineering, hemostasis, wound healing, drug delivery, and hemoperfusion. However, despite the wealth of studies on chitin/chitosan, there exists a notable trend of homogeneity in research, which could hinder the comprehensive development of these biomaterials. This review, taking a clinician's perspective, identifies current research gaps and medical challenges yet to be addressed, aiming to pave the way for a more sustainable future in chitin/chitosan research and application.
Collapse
Affiliation(s)
- Lanxin Mu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan 430071, China; Southwest Hospital of Third Military Medical University (Army Medical University), Department of Plastic Surgery, Chongqing 400038, China
| | - Liqin Wu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan 430071, China
| | - Shuangquan Wu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan 430071, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan 430071, China.
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan 430071, China.
| |
Collapse
|
43
|
Morais EA, Martins EMDN, Oliveira JADC, Melo EM, Mattos MS, Kraemer LR, Gomes DA, de Goes AM, Russo RC. Nanoformulated CHO-rPb27 vaccine enhances immunity and controls infection, mitigating lung inflammation and dysfunction during experimental Paracoccidioidomycosis in mice. Int J Biol Macromol 2024; 281:136261. [PMID: 39366607 DOI: 10.1016/j.ijbiomac.2024.136261] [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: 01/25/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Paracoccidioidomycosis (PCM) is a systemic mycosis caused by the dimorphic fungus within the genus Paracoccidioides, particularly Paracoccidioides brasiliensis. The traditional approach to treating this pulmonary infection involves prolonged therapy periods, ranging from weeks to years, often resulting in a notable frequency of disease relapse. Nanotechnology has emerged as a promising avenue for developing novel antifungal therapies and effective vaccines. This is attributed to its capability to facilitate targeted drug and antigen delivery, thereby mitigating toxicity and treatment expenses. This study investigates the synergistic properties of the CHO-rPb27 vaccine nanoformulation against experimental PCM. The therapeutic efficacy of CHO-rPb27 treatment is juxtaposed with the prophylactic protocol. Our findings demonstrate that both protocols effectively control P. brasiliensis pulmonary infection by eliciting a robust cellular and humoral immune response. This response attenuates chronic tissue damage and mitigates pulmonary mechanical dysfunction in mice.
Collapse
Affiliation(s)
- Elis Araujo Morais
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Estefania Mara do Nascimento Martins
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratory of Carbon Nanostructures Chemistry, National Commission for Nuclear Energy - Nuclear Technology Development Center (CDTN/CNEN), Belo Horizonte, Brazil
| | - Junnia Alvarenga de Carvalho Oliveira
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Eliza Mathias Melo
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus Silvério Mattos
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Rocha Kraemer
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Dawidson Assis Gomes
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alfredo Miranda de Goes
- Laboratory of Cellular and Molecular Immunology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
44
|
Ribeiro ARS, Neuper T, Horejs-Hoeck J. The Role of STING-Mediated Activation of Dendritic Cells in Cancer Immunotherapy. Int J Nanomedicine 2024; 19:10685-10697. [PMID: 39464674 PMCID: PMC11512692 DOI: 10.2147/ijn.s477320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
The signaling pathway that comprises cyclic guanosine monophosphate-adenosine monophosphate (cGAMP or GMP-AMP) synthase (cGAS) and Stimulator of Interferon Genes (STING) is emerging as a druggable target for immunotherapy, with tumor-resident dendritic cells (DC) playing a critical role in mediating its effects. The STING receptor is part of the DNA-sensing cellular machinery, that can trigger the secretion of pro-inflammatory mediators, priming effector T cells and initiating specific antitumor responses. Yet, recent studies have highlighted the dual role of STING activation in the context of cancer: STING can either promote antitumor responses or enhance tumor progression. This dichotomy often depends on the cell type in which cGAS-STING signaling is induced and the activation mode, namely acute versus chronic. Of note, STING activation at the DC level appears to be particularly important for tumor eradication. This review outlines the contribution of the different conventional and plasmacytoid DC subsets and describes the mechanisms underlying STING-mediated activation of DCs in cancer. We further highlight how the STING pathway plays an intricate role in modulating the function of DCs embedded in tumor tissue. Additionally, we discuss the strategies being employed to harness STING activation for cancer treatment, such as the development of synthetic agonists and nano-based delivery systems, spotlighting the current techniques used to prompt STING engagement specifically in DCs.
Collapse
Affiliation(s)
- Ana R S Ribeiro
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
| | - Theresa Neuper
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
- Center for Tumor biology and Immunology (CTBI), Salzburg, 5020, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
- Center for Tumor biology and Immunology (CTBI), Salzburg, 5020, Austria
| |
Collapse
|
45
|
Yu R, Jin L, Song Z, Jiao L, Wang Z, Zhou Y, Ma Y, Guan S, Zhang Z, Wang D, Liu H, Sun Y, Zhang S, Cai T, Sun H, Qiu Y, Miao J, Liu Z. A General Strategy toward Self-assembled Nanovaccine Based on Cationic Lentinan to Induce Potent Humoral and Cellular Immune Responses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402792. [PMID: 38940386 DOI: 10.1002/smll.202402792] [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: 04/08/2024] [Revised: 06/19/2024] [Indexed: 06/29/2024]
Abstract
Adjuvants play a critical role in the induction of effective immune responses by vaccines. Here, a self-assembling nanovaccine platform that integrates adjuvant functions into the delivery vehicle is prepared. Cationic Lentinan (CLNT) is mixed with ovalbumin (OVA) to obtain a self-assembling nanovaccine (CLNTO nanovaccine), which induces the uptake and maturation of bone marrow dendritic cells (BMDCs) via the toll-like receptors 2/4 (TLR2/4) to produce effective antigen cross-presentation. CLNTO nanovaccines target lymph nodes (LNs) and induce a robust OVA-specific immune response via TLR and tumor necrosis factor (TNF) signaling pathways, retinoic acid-inducible gene I (RIG-I) receptor, and cytokine-cytokine receptor interactions. In addition, CLNTO nanovaccines are found that promote the activation of follicular helper T (Tfh) cells and induce the differentiation of germinal center (GC) B cells into memory B cells and plasma cells, thereby enhancing the immune response. Vaccination with CLNTO nanovaccine significantly inhibits the growth of ovalbumin (OVA)-expressing B16 melanoma cell (B16-OVA) tumors, indicating its great potential for cancer immunotherapy. Therefore, this study presents a simple, safe, and effective self-assembling nanovaccine that induces helper T cell 1 (Th1) and helper T cell (Th2) immune responses, making it an effective vaccine delivery system.
Collapse
Affiliation(s)
- Ruihong Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Lan Jin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Zuchen Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Lina Jiao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Zheng Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Yantong Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Yan Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Sumei Guan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Zhimin Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Deyun Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Huina Liu
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315000, P. R. China
| | - Yuechao Sun
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315000, P. R. China
| | - Shun Zhang
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315000, P. R. China
| | - Ting Cai
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315000, P. R. China
| | - Haifeng Sun
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Yawei Qiu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Jinfeng Miao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
| | - Zhenguang Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, P. R. China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315000, P. R. China
| |
Collapse
|
46
|
Qi W, Yu Y, Yang C, Wang X, Jiang Y, Zhang L, Yu Z. Nanospheres as the delivery vehicle: novel application of Toxoplasma gondii ribosomal protein S2 in PLGA and chitosan nanospheres against acute toxoplasmosis. Front Immunol 2024; 15:1475280. [PMID: 39416787 PMCID: PMC11480959 DOI: 10.3389/fimmu.2024.1475280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Toxoplasma gondii (T. gondii) is a zoonotic disease that poses great harm to humans and animals. So far, no effective T. gondii vaccine has been developed to provide fully protection against such parasites. Recently, numerous researches have focused on the use of poly-lactic-co-glycolic acid (PLGA) and chitosan (CS) for the vaccines against T. gondii infections. In this study, we employed PLGA and CS as the vehicles for T. gondii ribosome protein (TgRPS2) delivery. TgRPS2-PLGA and TgRPS2-CS nanospheres were synthesized by double emulsion solvent evaporation and ionic gelation technique as the nano vaccines. Before immunization in animals, the release efficacy and toxicity of the synthesized nanospheres were evaluated in vitro. Then, ICR mice were immunized intramuscularly, and immune protections of the synthesized nanospheres were assessed. The results showed that TgRPS2-PLGA and TgRPS2-CS nanospheres could induce higher levels of IgG and cytokines, activate dendritic cells, and promote the expression of histocompatibility complexes. The splenic lymphocyte proliferation and the enhancement in the proportion of CD4+ and CD8+ T lymphocytes were also observed in immunized animals. In addition, two types of nanospheres could significantly inhabit the replications of T. gondii in cardiac muscles and spleen tissues. All these obtained results in this study demonstrated that the TgRPS2 protein delivered by PLGA or CS nanospheres provided satisfactory immunoprotective effects in resisting T. gondii, and such formulations illustrated potential as prospective preventive agents for toxoplasmosis.
Collapse
Affiliation(s)
- WeiYu Qi
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - YouLi Yu
- Institute of Animal Science, Ningxia Academy of Agricultural and Forestry Science, Yinchuan, China
| | - ChenChen Yang
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - XiaoJuan Wang
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - YuChen Jiang
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - Li Zhang
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - ZhengQing Yu
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| |
Collapse
|
47
|
Torres Quintas S, Canha-Borges A, Oliveira MJ, Sarmento B, Castro F. Special Issue: Nanotherapeutics in Women's Health Emerging Nanotechnologies for Triple-Negative Breast Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300666. [PMID: 36978237 DOI: 10.1002/smll.202300666] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/03/2023] [Indexed: 06/18/2023]
Abstract
Breast cancer appears as the major cause of cancer-related deaths in women, with more than 2 260 000 cases reported worldwide in 2020, resulting in 684 996 deaths. Triple-negative breast cancer (TNBC), characterized by the absence of estrogen, progesterone, and human epidermal growth factor type 2 receptors, represents ≈20% of all breast cancers. TNBC has a highly aggressive clinical course and is more prevalent in younger women. The standard therapy for advanced TNBC is chemotherapy, but responses are often short-lived, with high rate of relapse. The lack of therapeutic targets and the limited therapeutic options confer to individuals suffering from TNBC the poorest prognosis among breast cancer patients, remaining a major clinical challenge. In recent years, advances in cancer nanomedicine provided innovative therapeutic options, as nanoformulations play an important role in overcoming the shortcomings left by conventional therapies: payload degradation and its low solubility, stability, and circulating half-life, and difficulties regarding biodistribution due to physiological and biological barriers. In this integrative review, the recent advances in the nanomedicine field for TNBC treatment, including the novel nanoparticle-, exosome-, and hybrid-based therapeutic formulations are summarized and their drawbacks and challenges are discussed for future clinical applications.
Collapse
Affiliation(s)
- Sofia Torres Quintas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ana Canha-Borges
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Maria José Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- IUCS-CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116, Gandra, Portugal
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
| |
Collapse
|
48
|
Ren M, Abdullah SW, Pei C, Guo H, Sun S. Use of virus-like particles and nanoparticle-based vaccines for combating picornavirus infections. Vet Res 2024; 55:128. [PMID: 39350170 PMCID: PMC11443892 DOI: 10.1186/s13567-024-01383-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
Picornaviridae are non-enveloped ssRNA viruses that cause diseases such as poliomyelitis, hand-foot-and-mouth disease (HFMD), hepatitis A, encephalitis, myocarditis, and foot-and-mouth disease (FMD). Virus-like particles (VLPs) vaccines mainly comprise particles formed through the self-assembly of viral capsid proteins (for enveloped viruses, envelope proteins are also an option). They do not contain the viral genome. On the other hand, the nanoparticles vaccine (NPs) is mainly composed of self-assembling biological proteins or nanomaterials, with viral antigens displayed on the surface. The presentation of viral antigens on these particles in a repetitive array can elicit a strong immune response in animals. VLPs and NPs can be powerful platforms for multivalent antigen presentation. This review summarises the development of virus-like particle vaccines (VLPs) and nanoparticle vaccines (NPs) against picornaviruses. By detailing the progress made in the fight against various picornaviruses such as poliovirus (PV), foot-and-mouth disease virus (FMDV), enterovirus (EV), Senecavirus A (SVA), and encephalomyocarditis virus (EMCV), we in turn highlight the significant strides made in vaccine technology. These advancements include diverse construction methods, expression systems, elicited immune responses, and the use of various adjuvants. We see promising prospects for the continued development and optimisation of VLPs and NPs vaccines. Future research should focus on enhancing these vaccines' immunogenicity, stability, and delivery methods. Moreover, expanding our understanding of the interplay between these vaccines and the immune system will be crucial. We hope these insights will inspire and guide fellow researchers in the ongoing quest to combat picornavirus infections more effectively.
Collapse
Affiliation(s)
- Mei Ren
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gembloux Agro-Biotech, University of Liege, Gembloux, Belgium
| | - Sahibzada Waheed Abdullah
- Livestock and dairy development department peshawar, Government of Khyber Pakhtunkhwa, Peshawar, Pakistan
| | - Chenchen Pei
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, CollegeofVeterinaryMedicine, Lanzhou UniversityLanzhou Veterinary Research InstituteChinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
| |
Collapse
|
49
|
Wang Y, Liu C, Fang C, Peng Q, Qin W, Yan X, Zhang K. Engineered Cancer Nanovaccines: A New Frontier in Cancer Therapy. NANO-MICRO LETTERS 2024; 17:30. [PMID: 39347944 PMCID: PMC11442722 DOI: 10.1007/s40820-024-01533-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/08/2024] [Indexed: 10/01/2024]
Abstract
Vaccinations are essential for preventing and treating disease, especially cancer nanovaccines, which have gained considerable interest recently for their strong anti-tumor immune capabilities. Vaccines can prompt the immune system to generate antibodies and activate various immune cells, leading to a response against tumor tissues and reducing the negative effects and recurrence risks of traditional chemotherapy and surgery. To enhance the flexibility and targeting of vaccines, nanovaccines utilize nanotechnology to encapsulate or carry antigens at the nanoscale level, enabling more controlled and precise drug delivery to enhance immune responses. Cancer nanovaccines function by encapsulating tumor-specific antigens or tumor-associated antigens within nanomaterials. The small size of these nanomaterials allows for precise targeting of T cells, dendritic cells, or cancer cells, thereby eliciting a more potent anti-tumor response. In this paper, we focus on the classification of carriers for cancer nanovaccines, the roles of different target cells, and clinically tested cancer nanovaccines, discussing strategies for effectively inducing cytotoxic T lymphocytes responses and optimizing antigen presentation, while also looking ahead to the translational challenges of moving from animal experiments to clinical trials.
Collapse
Affiliation(s)
- Yijie Wang
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, People's Republic of China
| | - Congrui Liu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, People's Republic of China
| | - Chao Fang
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, People's Republic of China
| | - Qiuxia Peng
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, People's Republic of China
- Department of Stomatology and Central Laboratory, School of Medicine, Shanghai Tenth People's Hospital, Tongji University, NO. 301 Yan-Chang-Zhong Road, Shanghai, 200072, People's Republic of China
| | - Wen Qin
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, People's Republic of China
| | - Xuebing Yan
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, No.2, Bayi West Road, Suining, Xu Zhou, 221000, Jiangsu Province, People's Republic of China.
| | - Kun Zhang
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, People's Republic of China.
| |
Collapse
|
50
|
Liu Y, Wu Y, Li Z, Wan D, Pan J. Targeted Drug Delivery Strategies for the Treatment of Hepatocellular Carcinoma. Molecules 2024; 29:4405. [PMID: 39339402 PMCID: PMC11434448 DOI: 10.3390/molecules29184405] [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: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent malignant tumors, exhibiting a high incidence rate that presents a substantial threat to human health. The use of sorafenib and lenvatinib, commonly employed as single-agent targeted inhibitors, complicates the treatment process due to the absence of definitive targeting. Nevertheless, the advent of nanotechnology has injected new optimism into the domain of liver cancer therapy. Nanocarriers equipped with active targeting or passive targeting mechanisms have demonstrated the capability to deliver drugs to tumor cells with high efficiency. This approach not only facilitates precise delivery to the affected site but also enables targeted drug release, thereby enhancing therapeutic efficacy. As medical technology progresses, there is an increasing call for innovative treatment modalities, including novel chemotherapeutic agents, gene therapy, phototherapy, immunotherapy, and combinatorial treatments for HCC. These emerging therapies are anticipated to yield improved clinical outcomes for patients, while minimizing systemic toxicity and adverse effects. Consequently, the application of nanotechnology is poised to significantly improve HCC treatment. This review focused on targeted strategies for HCC and the application of nanotechnology in this area.
Collapse
Affiliation(s)
- Yonghui Liu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Yanan Wu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Zijian Li
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Dong Wan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Jie Pan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| |
Collapse
|