1
|
Liu Q, Huo X, Wang P, Zhao F, Yuan G, Yang C, Su J. Lactobacillus casei displaying MCP2α and FlaC delivered by PLA microspheres effectively enhances the immune protection of largemouth bass (Micropterus salmoides) against LMBV infection. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109870. [PMID: 39218416 DOI: 10.1016/j.fsi.2024.109870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Largemouth bass ranavirus (LMBV) seriously affects the development of largemouth bass (Micropterus salmoides) industry and causes huge economic losses. Oral vaccine can be a promising method for viral disease precaution. In this study, MCP2α was identified as a valuable epitope region superior to MCP and MCP2 of LMBV by neutralizing antibody experiments. Then, recombinant Lactobacillus casei expressing the fusion protein MCP2αC (MCP2α as antigen, C represents flagellin C from Aeromonas hydrophila as adjuvant) on surface was constructed and verified. Further, PLA microsphere vaccine loading recombinant MCP2αC L. casei was prepared. The PLA microspheres vaccine were observed by scanning electron microscopy and showed a smooth, regular spherical surface with a particle size distribution between 100 and 200 μm. Furthermore, we evaluated the tolerance of PLA-MCP2αC vaccine in simulated gastric fluid and simulated intestinal fluid, and the results showed that PLA-MCP2αC can effectively resist the gastrointestinal environment. Moreover, the protective effect of PLA-MCP2αC against LMBV was evaluated after oral immunization and LMBV challenge. The results showed that PLA-MCP2αC effectively up-regulated the activity of serum biochemical enzymes (T-SOD, T-AOC, LZM, complement C3) and induced the mRNA expression of representative immune genes (IL-1β, TNF-α, IFN-γ, MHC-IIα, Mx, IgM) in spleen and head kidney tissues. The survival rate of largemouth bass vaccinated with PLA-MCP2αC increased from 24 % to 68 %. Meanwhile, PLA-MCP2αC inhibited the LMBV burden in spleen, head kidney and liver tissues and attenuated tissue damage in spleen. These results suggested that PLA-MCP2αC can be used as a candidate oral vaccine against LMBV infection in aquaculture.
Collapse
Affiliation(s)
- Qian Liu
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Xingchen Huo
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Pengxu Wang
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Fengxia Zhao
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Gailing Yuan
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chunrong Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianguo Su
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
2
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
- Ziyi Bai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
3
|
Wu J, Wang X, Wang Y, Xun Z, Li S. Application of PLGA in Tumor Immunotherapy. Polymers (Basel) 2024; 16:1253. [PMID: 38732722 PMCID: PMC11085488 DOI: 10.3390/polym16091253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
Biodegradable polymers have been extensively researched in the field of biomedicine. Polylactic-co-glycolic acid (PLGA), a biodegradable polymer material, has been widely used in drug delivery systems and has shown great potential in various medical fields, including vaccines, tissue engineering such as bone regeneration and wound healing, and 3D printing. Cancer, a group of diseases with high mortality rates worldwide, has recently garnered significant attention in the field of immune therapy research. In recent years, there has been growing interest in the delivery function of PLGA in tumor immunotherapy. In tumor immunotherapy, PLGA can serve as a carrier to load antigens on its surface, thereby enhancing the immune system's ability to attack tumor cells. Additionally, PLGA can be used to formulate tumor vaccines and immunoadjuvants, thereby enhancing the efficacy of tumor immunotherapy. PLGA nanoparticles (NPs) can also enhance the effectiveness of tumor immunotherapy by regulating the activity and differentiation of immune cells, and by improving the expression and presentation of tumor antigens. Furthermore, due to the diverse physical properties and surface modifications of PLGA, it has a wider range of potential applications in tumor immunotherapy through the loading of various types of drugs or other innovative substances. We aim to highlight the recent advances and challenges of plga in the field of oncology therapy to stimulate further research and development of innovative PLGA-based approaches, and more effective and personalized cancer therapies.
Collapse
Affiliation(s)
- Jiashuai Wu
- Innovation Institute, China Medical University, Shenyang 110122, China; (J.W.); (X.W.)
| | - Xiaopeng Wang
- Innovation Institute, China Medical University, Shenyang 110122, China; (J.W.); (X.W.)
| | - Yunduan Wang
- School of Intelligent Medicine, China Medical University, Shenyang 110122, China;
| | - Zhe Xun
- Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Health Science Institute, China Medical University, Shenyang 110122, China
| | - Shuo Li
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
4
|
Ali M, Mohd Noor SNF, Mohamad H, Ullah F, Javed F, Abdul Hamid ZA. Advances in guided bone regeneration membranes: a comprehensive review of materials and techniques. Biomed Phys Eng Express 2024; 10:032003. [PMID: 38224615 DOI: 10.1088/2057-1976/ad1e75] [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: 06/06/2023] [Accepted: 01/15/2024] [Indexed: 01/17/2024]
Abstract
Guided tissue/bone regeneration (GTR/GBR) is a widely used technique in dentistry to facilitate the regeneration of damaged bone and tissue, which involves guiding materials that eventually degrade, allowing newly created tissue to take its place. This comprehensive review the evolution of biomaterials for guided bone regeneration that showcases a progressive shift from non-resorbable to highly biocompatible and bioactive materials, allowing for more effective and predictable bone regeneration. The evolution of biomaterials for guided bone regeneration GTR/GBR has marked a significant progression in regenerative dentistry and maxillofacial surgery. Biomaterials used in GBR have evolved over time to enhance biocompatibility, bioactivity, and efficacy in promoting bone growth and integration. This review also probes into several promising fabrication techniques like electrospinning and latest 3D printing fabrication techniques, which have shown potential in enhancing tissue and bone regeneration processes. Further, the challenges and future direction of GTR/GBR are explored and discussed.
Collapse
Affiliation(s)
- Mohammed Ali
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| | - Siti Noor Fazliah Mohd Noor
- Dental Stimulation and Virtual Learning, Research Excellence Consortium, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Hasmaliza Mohamad
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| | - Faheem Ullah
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
- Department of Biological Sciences, Biopolymer Research Centre (BRC), National University of Medical Sciences, 46000, Rawalpindi, Pakistan
| | - Fatima Javed
- Department of Chemistry, Shaheed Benazir Butto Women University Peshawar, Charsadda Road Laramma, 25000, Peshawar, Pakistan
| | - Zuratul Ain Abdul Hamid
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| |
Collapse
|
5
|
Freitas R, Ferreira E, Miranda A, Ferreira D, Relvas-Santos M, Castro F, Santos B, Gonçalves M, Quintas S, Peixoto A, Palmeira C, Silva AMN, Santos LL, Oliveira MJ, Sarmento B, Ferreira JA. Targeted and Self-Adjuvated Nanoglycovaccine Candidate for Cancer Immunotherapy. ACS NANO 2024; 18:10088-10103. [PMID: 38535625 DOI: 10.1021/acsnano.3c12487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Advanced-stage solid primary tumors and metastases often express mucin 16 (MUC16), carrying immature glycans such as the Tn antigen, resulting in specific glycoproteoforms not found in healthy human tissues. This presents a valuable approach for designing targeted therapeutics, including cancer glycovaccines, which could potentially promote antigen recognition and foster the immune response to control disease spread and prevent relapse. In this study, we describe an adjuvant-free poly(lactic-co-glycolic acid) (PLGA)-based nanoglycoantigen delivery approach that outperforms conventional methods by eliminating the need for protein carriers while exhibiting targeted and adjuvant properties. To achieve this, we synthesized a library of MUC16-Tn glycoepitopes through single-pot enzymatic glycosylation, which were then stably engrafted onto the surface of PLGA nanoparticles, generating multivalent constructs that better represent cancer molecular heterogeneity. These glycoconstructs demonstrated affinity for Macrophage Galactose-type Lectin (MGL) receptor, known to be highly expressed by immature antigen-presenting cells, enabling precise targeting of immune cells. Moreover, the glycopeptide-grafted nanovaccine candidate displayed minimal cytotoxicity and induced the activation of dendritic cells in vitro, even in the absence of an adjuvant. In vivo, the formulated nanovaccine candidate was also nontoxic and elicited the production of IgG specifically targeting MUC16 and MUC16-Tn glycoproteoforms in cancer cells and tumors, offering potential for precise cancer targeting, including targeted immunotherapies.
Collapse
Affiliation(s)
- Rui Freitas
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Eduardo Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
| | - Andreia Miranda
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Dylan Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Flávia Castro
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Beatriz Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Martina Gonçalves
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Sofia Quintas
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Carlos Palmeira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- Immunology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
- Health School of University Fernando Pessoa, 4249-004 Porto, Portugal
| | - André M N Silva
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
- GlycoMatters Biotech, 4500-162 Espinho, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- GlycoMatters Biotech, 4500-162 Espinho, Portugal
- Department of Surgical Oncology, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - Maria José Oliveira
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- IUCS-CESPU, 4585-116 Gandra, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, Research Center of IPO-Porto (CI-IPOP), 4200-072 Porto, Portugal
- RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), 4200-072 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- GlycoMatters Biotech, 4500-162 Espinho, Portugal
| |
Collapse
|
6
|
Piva-Amaral R, Augusto Pires de Souza G, Carlos Vilela Vieira Júnior J, Fróes Goulart de Castro R, Permagnani Gozzi W, Pereira Lima Neto S, Cauvilla Dos Santos AL, Pavani Cassiano H, Christine Ferreira da Silva L, Dias Novaes R, Santos Abrahão J, Ervolino de Oliveira C, de Mello Silva B, de Paula Costa G, Cosme Cotta Malaquias L, Felipe Leomil Coelho L. Bovine serum albumin nanoparticles containing Poly (I:C) can enhance the neutralizing antibody response induced by envelope protein of Orthoflavivirus zikaense. Int Immunopharmacol 2024; 128:111523. [PMID: 38219440 DOI: 10.1016/j.intimp.2024.111523] [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: 11/21/2023] [Revised: 12/21/2023] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Since the Orthoflavivirus zikaense (ZIKV) has been considered a risk for Zika congenital syndrome development, developing a safe and effective vaccine has become a high priority. Numerous research groups have developed strategies to prevent ZIKV infection and have identified the domain III of the ZIKV envelope protein (zEDIII) as a promising target. Subunit antigens are often poorly immunogenic, necessitating the use of adjuvants and/or delivery systems to induce optimal immune responses. The subject of nanotechnology has substantial expansion in recent years in terms of research and applications. Nanoparticles could be used as drug delivery systems and to increase the immunogenicity and stability of a given antigen. This work aims to characterize and validate the potential of a vaccine formulation composed of domain zEDIII and bovine serum albumin nanoparticles containing polyinosinic-polycytidylic acid (NPPI). NPPI were uptake in vitro by immature bone marrow dendritic cells and histological analysis of the skin of mice treated with NPPI showed an increase in cellularity. Immunization assay showed that mice immunized with zEDIII in the presence of NPPI produced neutralizing antibodies. Through the passive transfer of sera from immunized mice to ZIKV-infected neonatal mice, it was demonstrated that these antibodies provide protection, mitigating weight loss, clinical or neurological signs induced by infection, and significantly increased survival rates. Protection was further substantiated by the reduction in the number of viable infectious ZIKV, as well as a decrease in inflammatory cytokines and tissue alterations in the brains of infected mice. Taken together, data presented in this study shows that NPPI + zEDIII is a promising vaccine candidate for ZIKV.
Collapse
Affiliation(s)
- Raíne Piva-Amaral
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil.
| | - Gabriel Augusto Pires de Souza
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil; Laboratório de Vírus, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Brazil
| | - João Carlos Vilela Vieira Júnior
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil
| | - Renato Fróes Goulart de Castro
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil
| | - William Permagnani Gozzi
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil
| | - Sergio Pereira Lima Neto
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil
| | - Ana Luisa Cauvilla Dos Santos
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil
| | - Helena Pavani Cassiano
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil
| | | | - Romulo Dias Novaes
- Instituto de Ciências Biomédicas, Departamento de Biologia Estrutural, Universidade Federal de Alfenas, 37130-001 Minas Gerais, Brazil
| | - Jônatas Santos Abrahão
- Laboratório de Vírus, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Brazil
| | - Carine Ervolino de Oliveira
- Instituto de Ciências Biomédicas, Departamento de Patologia e Parasitologia, Universidade Federal de Alfenas, 37130-001 Minas Gerais, Brazil
| | - Breno de Mello Silva
- Núcleo de Pesquisas em Ciências Biológicas, NUPEB, Universidade Federal de Ouro Preto, Ouro Preto 35400-000, Brazil
| | - Guilherme de Paula Costa
- Núcleo de Pesquisas em Ciências Biológicas, NUPEB, Universidade Federal de Ouro Preto, Ouro Preto 35400-000, Brazil
| | - Luiz Cosme Cotta Malaquias
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil
| | - Luiz Felipe Leomil Coelho
- Laboratório de Vacinas, Departamento de Microbiologia e Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas 37130-001, Brazil.
| |
Collapse
|
7
|
Sun Z, Zhao H, Ma L, Shi Y, Ji M, Sun X, Ma D, Zhou W, Huang T, Zhang D. The quest for nanoparticle-powered vaccines in cancer immunotherapy. J Nanobiotechnology 2024; 22:61. [PMID: 38355548 PMCID: PMC10865557 DOI: 10.1186/s12951-024-02311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Despite recent advancements in cancer treatment, this disease still poses a serious threat to public health. Vaccines play an important role in preventing illness by preparing the body's adaptive and innate immune responses to combat diseases. As our understanding of malignancies and their connection to the immune system improves, there has been a growing interest in priming the immune system to fight malignancies more effectively and comprehensively. One promising approach involves utilizing nanoparticle systems for antigen delivery, which has been shown to potentiate immune responses as vaccines and/or adjuvants. In this review, we comprehensively summarized the immunological mechanisms of cancer vaccines while focusing specifically on the recent applications of various types of nanoparticles in the field of cancer immunotherapy. By exploring these recent breakthroughs, we hope to identify significant challenges and obstacles in making nanoparticle-based vaccines and adjuvants feasible for clinical application. This review serves to assess recent breakthroughs in nanoparticle-based cancer vaccinations and shed light on their prospects and potential barriers. By doing so, we aim to inspire future immunotherapies for cancer that harness the potential of nanotechnology to deliver more effective and targeted treatments.
Collapse
Affiliation(s)
- Zhe Sun
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hui Zhao
- Department of Endodontics, East Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Li Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yanli Shi
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Mei Ji
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xiaodong Sun
- Department of Endodontics, Gaoxin Branch of Jinan Stomatological Hospital, Jinan, 250000, Shandong, China
| | - Dan Ma
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Wei Zhou
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Tao Huang
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Dongsheng Zhang
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
8
|
Buchard A, Davidson MG, Gobius du Sart G, Jones MD, Kociok-Köhn G, McCormick SN, McKeown P. Unexpected Periodicity in Cationic Group 5 Initiators for the Ring-Opening Polymerization of Lactones. Inorg Chem 2024; 63:27-38. [PMID: 38118120 PMCID: PMC10777398 DOI: 10.1021/acs.inorgchem.3c03854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/23/2023] [Accepted: 12/04/2023] [Indexed: 12/22/2023]
Abstract
ε-Caprolactone (ε-CL) adducts of cationic, amine tris(phenolate)-supported niobium(V) and tantalum(V) ethoxides initiate the ring-opening polymerization of lactones. The Ta(V) species prepared and applied catalytically herein exhibits higher activity in the ring-opening polymerization (ROP) of ε-caprolactone than the previously reported, isostructural Nb(V) complex, contradicting literature comparisons of Nb(V)- and Ta(V)-based protocols. Both systems also initiate the ROP of δ-valerolactone and rac-β-butyrolactone, kinetic studies confirming retention of higher activity by the Ta congener. Polymerizations of rac-β-butyrolactone and δ-valerolactone were previously unrealized under Group V- or Ta-mediated conditions, respectively, although the former has afforded only low molecular weight, cyclic poly-3-hydroxybutyrate. Cationic ethoxo-Nb(V) and -Ta(V) δ-valerolactone adducts are also reported, demonstrating the facility of δ-valerolactone as a ligand and the generality of the synthetic method. Both δ-valerolactone-bearing complexes initiate the ROP of ε-caprolactone, δ-valerolactone, and rac-β-butyrolactone. Accordingly, we have elucidated trends in reactivity and investigated the initiation mechanism for such systems, the insertion event being predicated upon intramolecular nucleophilic attack on the coordinated lactone by the adjacent alkoxide moiety. This mechanism enables quantitative, stoichiometric installation of a single monomer residue distinct from the bulk of the polymer chain, and permits modification of polymer properties via both manipulation of the molecular architecture and tuning of the polymerization kinetics, and thus dispersity, through hitherto inaccessible independent control of the initiation event.
Collapse
Affiliation(s)
- Antoine Buchard
- Institute
for Sustainability, University of Bath, Bath BA2 7AY, United Kingdom
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Matthew G. Davidson
- Institute
for Sustainability, University of Bath, Bath BA2 7AY, United Kingdom
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | | | - Matthew D. Jones
- Institute
for Sustainability, University of Bath, Bath BA2 7AY, United Kingdom
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Gabriele Kociok-Köhn
- Material
and Chemical Characterization and Analysis Facility (MC), University of Bath, Bath BA2 7AY, United Kingdom
| | - Strachan N. McCormick
- Institute
for Sustainability, University of Bath, Bath BA2 7AY, United Kingdom
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Paul McKeown
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
| |
Collapse
|
9
|
Zhang Y, Zhang Z, Liu Y, Cai D, Gu J, Sun D. Differential Mobility Spectrometry-Tandem Mass Spectrometry with Multiple Ion Monitoring Coupled with in Source-Collision Induced Dissociation: A New Strategy for the Quantitative Analysis of Pharmaceutical Polymer Excipients in Rat Plasma. Molecules 2023; 28:4782. [PMID: 37375337 DOI: 10.3390/molecules28124782] [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: 05/16/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Polylactic acids (PLAs) are synthetic polymers composed of repeating lactic acid subunits. For their good biocompatibility, PLAs have been approved and widely applied as pharmaceutical excipients and scaffold materials. Liquid chromatography-tandem mass spectrometry is a powerful analytical tool not only for pharmaceutical ingredients but also for pharmaceutical excipients. However, the characterization of PLAs presents particular problems for mass spectrometry techniques. In addition to their high molecular weights and wide polydispersity, multiple charging and various adductions are intrinsic features of electrospray ionization. In the present study, a strategy combining of differential mobility spectrometry (DMS), multiple ion monitoring (MIM) and in-source collision-induced dissociation (in source-CID) has been developed and applied to the characterization and quantitation of PLAs in rat plasma. First, PLAs will be fragmented into characteristic fragment ions under high declustering potential in the ionization source. The specific fragment ions are then screened twice by quadrupoles to ensure a high signal intensity and low interference for mass spectrometry detection. Subsequently, DMS technique has been applied to further reduce the background noise. The appropriately chosen surrogate specific precursor ions could be utilized for the qualitative and quantitative analysis of PLAs, which provided results with the advantages of low endogenous interference, sufficient sensitivity and selectivity for bioassay. The linearity of the method was evaluated over the concentration range 3-100 μg/mL (r2 = 0.996) for PLA 20,000. The LC-DMS-MIM coupled with in source-CID strategy may contribute to the pharmaceutical studies of PLAs and the possible prospects of other pharmaceutical excipients.
Collapse
Affiliation(s)
- Yuyao Zhang
- Research Center for Drug Metabolism, School of Life Science, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhi Zhang
- Research Center for Drug Metabolism, School of Life Science, Jilin University, Changchun 130012, China
| | - Yingze Liu
- Research Center for Drug Metabolism, School of Life Science, Jilin University, Changchun 130012, China
| | - Deqi Cai
- Research Center for Drug Metabolism, School of Life Science, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jingkai Gu
- Research Center for Drug Metabolism, School of Life Science, Jilin University, Changchun 130012, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Dong Sun
- Research Center for Drug Metabolism, School of Life Science, Jilin University, Changchun 130012, China
| |
Collapse
|
10
|
Liu S, Yu JM, Gan YC, Qiu XZ, Gao ZC, Wang H, Chen SX, Xiong Y, Liu GH, Lin SE, McCarthy A, John JV, Wei DX, Hou HH. Biomimetic natural biomaterials for tissue engineering and regenerative medicine: new biosynthesis methods, recent advances, and emerging applications. Mil Med Res 2023; 10:16. [PMID: 36978167 PMCID: PMC10047482 DOI: 10.1186/s40779-023-00448-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/23/2023] [Indexed: 03/30/2023] Open
Abstract
Biomimetic materials have emerged as attractive and competitive alternatives for tissue engineering (TE) and regenerative medicine. In contrast to conventional biomaterials or synthetic materials, biomimetic scaffolds based on natural biomaterial can offer cells a broad spectrum of biochemical and biophysical cues that mimic the in vivo extracellular matrix (ECM). Additionally, such materials have mechanical adaptability, microstructure interconnectivity, and inherent bioactivity, making them ideal for the design of living implants for specific applications in TE and regenerative medicine. This paper provides an overview for recent progress of biomimetic natural biomaterials (BNBMs), including advances in their preparation, functionality, potential applications and future challenges. We highlight recent advances in the fabrication of BNBMs and outline general strategies for functionalizing and tailoring the BNBMs with various biological and physicochemical characteristics of native ECM. Moreover, we offer an overview of recent key advances in the functionalization and applications of versatile BNBMs for TE applications. Finally, we conclude by offering our perspective on open challenges and future developments in this rapidly-evolving field.
Collapse
Affiliation(s)
- Shuai Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900, China
| | - Jiang-Ming Yu
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Yan-Chang Gan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900, China
| | - Xiao-Zhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900, China
| | - Zhe-Chen Gao
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Huan Wang
- The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, Guangdong, China.
| | - Shi-Xuan Chen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325011, Zhejiang, China.
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Si-En Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Alec McCarthy
- Department of Functional Materials, Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Johnson V John
- Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68130, USA
| | - Dai-Xu Wei
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336, China.
- Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong, 643002, Sichuan, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710127, China.
| | - Hong-Hao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, The Fifth Affiliated Hospital, School of Basic Medical Science, Southern Medical University, Guangzhou, 510900, China.
| |
Collapse
|
11
|
Conde G, Aracati MF, Rodrigues LF, de Oliveira SL, da Costa CC, Charlie-Silva I, Ruiz TFR, Taboga SR, Belo MADA. Device implant based on poly (lactic acid) with vitamin E for vaccine delivery system in Tilapia: Study for biocompatibility and biodegradation. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2022; 3:100060. [DOI: 10.1016/j.fsirep.2022.100060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 12/30/2022] Open
|
12
|
Monge C, Ayad C, Paris AL, Rovera R, Colomb E, Verrier B. Mucosal Adjuvants Delivered by a Mucoadhesive Patch for Sublingual Administration of Subunit Vaccines. Int J Mol Sci 2022; 23:13440. [PMID: 36362224 PMCID: PMC9655718 DOI: 10.3390/ijms232113440] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 11/19/2023] Open
Abstract
Among mucosal administration routes for vaccines, the sublingual route has been proven capable of inducing a potent systemic and mucosal immune response. However, the absence of a simple and compliant delivery system and the lack of robust mucosal adjuvants impede the development of sublingual vaccines. Here, we describe a mucoadhesive patch made of a layer-by-layer assembly of polysaccharides, chitosan, and hyaluronic acid. The mucoadhesive patch was covered by adjuvanted nanoparticles carrying viral proteins. We showed that the nanoparticles effectively cross the outer layers of the sublingual mucosa to reach the epithelium. Furthermore, the encapsulated adjuvants, 3M-052 and mifamurtide, targeting toll-like receptor (TLR) 7/8 and nucleotide-binding oligomerization domain-2 (NOD2), respectively, remain fully active after encapsulation into nanoparticles and exhibit a cytokine/chemokine signature similar to the mucosal gold-standard adjuvant, the cholera toxin. However, the particulate adjuvants induced more moderate levels of proinflammatory interleukin (IL)-6 and keratinocyte chemoattractant (KC), suggesting a controlled activation of the innate immune response.
Collapse
Affiliation(s)
- Claire Monge
- UMR 5305: Laboratoire de Biologie Tissulaire et d’Ingénierie Thérapeutique, Institut de Biologie et Chimie des Protéines, CNRS/Université Claude Bernard Lyon 1, 7 Passage du Vercors, 69007 Lyon, France
| | | | | | | | | | | |
Collapse
|
13
|
Tsachouridis K, Christodoulou E, Zamboulis A, Michopoulou A, Barmpalexis P, Bikiaris DN. Evaluation of poly(lactic acid)/ and poly(lactic-co-glycolic acid)/ poly(ethylene adipate) copolymers for the preparation of paclitaxel loaded drug nanoparticles. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
14
|
Recent approaches to mRNA vaccine delivery by lipid-based vectors prepared by continuous-flow microfluidic devices. Future Med Chem 2022; 14:1561-1581. [DOI: 10.4155/fmc-2022-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Advancements in nanotechnology have resulted in the introduction of several nonviral delivery vectors for the nontoxic, efficient delivery of encapsulated mRNA-based vaccines. Lipid- and polymer-based nanoparticles (NP) have proven to be the most potent delivery systems, providing increased delivery efficiency and protection of mRNA molecules from degradation. Here, the authors provide an overview of the recent studies carried out using lipid NPs and their functionalized forms, polymeric and lipid-polymer hybrid nanocarriers utilized mainly for the encapsulation of mRNAs for gene and immune therapeutic applications. A microfluidic system as a prevalent methodology for the preparation of NPs with continuous flow enables NP size tuning, rapid mixing and production reproducibility. Continuous-flow microfluidic devices for lipid and polymeric encapsulated RNA NP production are specifically reviewed.
Collapse
|
15
|
Zhang X, Cui H, Zhang W, Li Z, Gao J. Engineered tumor cell-derived vaccines against cancer: The art of combating poison with poison. Bioact Mater 2022; 22:491-517. [PMID: 36330160 PMCID: PMC9619151 DOI: 10.1016/j.bioactmat.2022.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 12/23/2022] Open
Abstract
Tumor vaccination is a promising approach for tumor immunotherapy because it presents high specificity and few side effects. However, tumor vaccines that contain only a single tumor antigen can allow immune system evasion by tumor variants. Tumor antigens are complex and heterogeneous, and identifying a single antigen that is uniformly expressed by tumor cells is challenging. Whole tumor cells can produce comprehensive antigens that trigger extensive tumor-specific immune responses. Therefore, tumor cells are an ideal source of antigens for tumor vaccines. A better understanding of tumor cell-derived vaccines and their characteristics, along with the development of new technologies for antigen delivery, can help improve vaccine design. In this review, we summarize the recent advances in tumor cell-derived vaccines in cancer immunotherapy and highlight the different types of engineered approaches, mechanisms, administration methods, and future perspectives. We discuss tumor cell-derived vaccines, including whole tumor cell components, extracellular vesicles, and cell membrane-encapsulated nanoparticles. Tumor cell-derived vaccines contain multiple tumor antigens and can induce extensive and potent tumor immune responses. However, they should be engineered to overcome limitations such as insufficient immunogenicity and weak targeting. The genetic and chemical engineering of tumor cell-derived vaccines can greatly enhance their targeting, intelligence, and functionality, thereby realizing stronger tumor immunotherapy effects. Further advances in materials science, biomedicine, and oncology can facilitate the clinical translation of tumor cell-derived vaccines.
Collapse
Affiliation(s)
- Xinyi Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Hengqing Cui
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Wenjun Zhang
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Corresponding author. Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China,Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China,Corresponding author. Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200444, China.
| |
Collapse
|
16
|
Maadani AM, Salahinejad E. Performance comparison of PLA- and PLGA-coated porous bioceramic scaffolds: Mechanical, biodegradability, bioactivity, delivery and biocompatibility assessments. J Control Release 2022; 351:1-7. [PMID: 36115555 DOI: 10.1016/j.jconrel.2022.09.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/10/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022]
Abstract
Bioceramics, particularly calcium phosphates, bioactive glasses, and crystalline silicates, are a principal group of biomaterials employed for the regeneration of damaged tissues and therapeutic delivery. The development of ceramic tissue engineering scaffolds with an appropriate combination of mechanical and biological properties is still one of the key challenges in this field. In this regard, the deposition of polymeric coatings on the scaffolds is a simple and effective approach to reinforce their functions. Among different polymers, the influences of biodegradable aliphatic polyester coatings, especially polylactic acid (PLA) and poly(lactic-co-glycolic acid) (PLGA), over the performance of the scaffolds have been investigated in numerous research. This review paper provides a comprehensive comparison of PLA- and PLGA-coated bioceramic scaffolds which are mainly employed in bone tissue engineering. It is concluded that both the polymers enhance the mechanical behaviors of the scaffolds, but control their biodegradability, bioactivity, and delivery kinetics, where PLA acts almost more influentially than PLGA in comparison. However, the response of biocompatibility to this surface treatment is condition-dependent and requires case-by-case experiments to be determined accurately.
Collapse
Affiliation(s)
- Amir Mohammad Maadani
- Faculty of Materials Science and Engineering, K.N. Toosi University of Technology, Tehran, Iran
| | - Erfan Salahinejad
- Faculty of Materials Science and Engineering, K.N. Toosi University of Technology, Tehran, Iran.
| |
Collapse
|
17
|
Flavonoid-based Polymeric Nanoparticles: A Promising Approach for Cancer and Diabetes Treatment. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Fan J, Jin S, Gilmartin L, Toth I, Hussein WM, Stephenson RJ. Advances in Infectious Disease Vaccine Adjuvants. Vaccines (Basel) 2022; 10:1120. [PMID: 35891284 PMCID: PMC9316175 DOI: 10.3390/vaccines10071120] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Vaccines are one of the most significant medical interventions in the fight against infectious diseases. Since their discovery by Edward Jenner in 1796, vaccines have reduced the worldwide transmission to eradication levels of infectious diseases, including smallpox, diphtheria, hepatitis, malaria, and influenza. However, the complexity of developing safe and effective vaccines remains a barrier for combating many more infectious diseases. Immune stimulants (or adjuvants) are an indispensable factor in vaccine development, especially for inactivated and subunit-based vaccines due to their decreased immunogenicity compared to whole pathogen vaccines. Adjuvants are widely diverse in structure; however, their overall function in vaccine constructs is the same: to enhance and/or prolong an immunological response. The potential for adverse effects as a result of adjuvant use, though, must be acknowledged and carefully managed. Understanding the specific mechanisms of adjuvant efficacy and safety is a key prerequisite for adjuvant use in vaccination. Therefore, rigorous pre-clinical and clinical research into adjuvant development is essential. Overall, the incorporation of adjuvants allows for greater opportunities in advancing vaccine development and the importance of immune stimulants drives the emergence of novel and more effective adjuvants. This article highlights recent advances in vaccine adjuvant development and provides detailed data from pre-clinical and clinical studies specific to infectious diseases. Future perspectives into vaccine adjuvant development are also highlighted.
Collapse
Affiliation(s)
- Jingyi Fan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Shengbin Jin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Lachlan Gilmartin
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| | - Rachel J. Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia; (J.F.); (S.J.); (L.G.); (I.T.); (W.M.H.)
| |
Collapse
|
19
|
Activation of Cellular Players in Adaptive Immunity via Exogenous Delivery of Tumor Cell Lysates. Pharmaceutics 2022; 14:pharmaceutics14071358. [PMID: 35890254 PMCID: PMC9316852 DOI: 10.3390/pharmaceutics14071358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 12/04/2022] Open
Abstract
Tumor cell lysates (TCLs) are a good immunogenic source of tumor-associated antigens. Since whole necrotic TCLs can enhance the maturation and antigen-presenting ability of dendritic cells (DCs), multiple strategies for the exogenous delivery of TCLs have been investigated as novel cancer immunotherapeutic solutions. The TCL-mediated induction of DC maturation and the subsequent immunological response could be improved by utilizing various material-based carriers. Enhanced antitumor immunity and cancer vaccination efficacy could be eventually achieved through the in vivo administration of TCLs. Therefore, (1) important engineering methodologies to prepare antigen-containing TCLs, (2) current therapeutic approaches using TCL-mediated DC activation, and (3) the significant sequential mechanism of DC-based signaling and stimulation in adaptive immunity are summarized in this review. More importantly, the recently reported developments in biomaterial-based exogenous TCL delivery platforms and co-delivery strategies with adjuvants for effective cancer vaccination and antitumor effects are emphasized.
Collapse
|
20
|
Lv S, Song K, Yen A, Peeler DJ, Nguyen DC, Olshefsky A, Sylvestre M, Srinivasan S, Stayton PS, Pun SH. Well-Defined Mannosylated Polymer for Peptide Vaccine Delivery with Enhanced Antitumor Immunity. Adv Healthc Mater 2022; 11:e2101651. [PMID: 34706166 PMCID: PMC9043035 DOI: 10.1002/adhm.202101651] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/19/2021] [Indexed: 12/28/2022]
Abstract
Peptide-based cancer vaccines offer production and safety advantages but have had limited clinical success due to their intrinsic instability, rapid clearance, and low cellular uptake. Nanoparticle-based delivery vehicles can improve the in vivo stability and cellular uptake of peptide antigens. Here, a well-defined, self-assembling mannosylated polymer is developed for anticancer peptide antigen delivery. The amphiphilic polymer is prepared by reversible addition-fragmentation chain transfer (RAFT) polymerization, and the peptide antigens are conjugated to the pH-sensitive hydrophobic block through the reversible disulfide linkage for selective release after cell entry. The polymer-peptide conjugates self-assemble into sub-100 nm micelles at physiological pH and dissociate at endosomal pH. The mannosylated micellar corona increases the accumulation of vaccine cargoes in the draining inguinal lymph nodes and facilitates nanoparticle uptake by professional antigen presenting cells. In vivo studies demonstrate that the mannosylated micelle formulation improves dendritic cell activation and enhances antigen-specific T cell responses, resulting in higher antitumor immunity in tumor-bearing mice compared to free peptide antigen. The mannosylated polymer is therefore a simple and promising platform for the delivery of peptide cancer vaccines.
Collapse
Affiliation(s)
- Shixian Lv
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Kefan Song
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Albert Yen
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - David J Peeler
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Dinh Chuong Nguyen
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Audrey Olshefsky
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Meilyn Sylvestre
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Selvi Srinivasan
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Patrick S Stayton
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Suzie H Pun
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
21
|
Espinar-Buitrago M, Muñoz-Fernández MA. New Approaches to Dendritic Cell-Based Therapeutic Vaccines Against HIV-1 Infection. Front Immunol 2022; 12:719664. [PMID: 35058917 PMCID: PMC8763680 DOI: 10.3389/fimmu.2021.719664] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Due to the success of combined antiretroviral therapy (cART) in recent years, the pathological outcome of Human Immunodeficiency Virus type 1 (HIV-1) infection has improved substantially, achieving undetectable viral loads in most cases. Nevertheless, the presence of a viral reservoir formed by latently infected cells results in patients having to maintain treatment for life. In the absence of effective eradication strategies against HIV-1, research efforts are focused on obtaining a cure. One of these approaches is the creation of therapeutic vaccines. In this sense, the most promising one up to now is based on the establishing of the immunological synapse between dendritic cells (DCs) and T lymphocytes (TL). DCs are one of the first cells of the immune system to encounter HIV-1 by acting as antigen presenting cells, bringing about the interaction between innate and adaptive immune responses mediated by TL. Furthermore, TL are the end effector, and their response capacity is essential in the adaptive elimination of cells infected by pathogens. In this review, we summarize the knowledge of the interaction between DCs with TL, as well as the characterization of the specific T-cell response against HIV-1 infection. The use of nanotechnology in the design and improvement of vaccines based on DCs has been researched and presented here with a special emphasis.
Collapse
Affiliation(s)
- Marisierra Espinar-Buitrago
- Section Head Immunology, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Ma Angeles Muñoz-Fernández
- Section Head Immunology, Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Spanish Human Immunodeficiency Virus- Hospital Gregorio Marañón (HIV-HGM) BioBank, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
22
|
Soares DCF, Poletto F, Eberhardt MJ, Domingues SC, De Sousa FB, Tebaldi ML. Polymer-hybrid nanosystems for antiviral applications: Current advances. Biomed Pharmacother 2022; 146:112249. [PMID: 34972632 DOI: 10.1016/j.biopha.2021.112249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 11/02/2022] Open
Abstract
The emergence of many new viruses in recent times has resulted in a significant scientific challenge for discovering drugs and vaccines that effectively treat and prevent viral diseases. Nanotechnology has opened doors to prevent the spread of several diseases, including those caused by viruses. Polymer-hybrid nanodevices are a class of nanotechnology platforms for biomedical applications that present synergistic properties among their components, with improved performance compared to conventional forms of therapy. Considering the growing interest in this emerging field and the promising technological advantages of polymer-hybrid nanodevices, this work presents the current status of these systems in the context of prevention and treatment of viral diseases. A brief description of the different types of polymer-hybrid nanodevices highlighting some peculiar characteristics such as their composition, biodistribution, delivery of antigens, and overall immune responses in systemic tissues are discussed. Finally, the work presents the future trends for new nanotechnological hybrid materials based on polymers and perspectives for clinical use.
Collapse
Affiliation(s)
| | - Fernanda Poletto
- Programa de Pós-Graduação em Química, Instituto de Química, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 91501-970, Brazil
| | - Marcelo J Eberhardt
- Programa de Pós-Graduação em Química, Instituto de Química, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 91501-970, Brazil
| | - Stephanie Calazans Domingues
- Laboratório de Bioengenharia - Universidade Federal de Itajubá (UNIFEI) - Campus Itabira, Itabira, MG 35903-087, Brazil
| | - Frederico B De Sousa
- Laboratório de Sistemas Poliméricos e Supramoleculares (LSPS) - Instituto de Física e Química, Universidade Federal de Itajubá (UNIFEI), Itajubá, MG 37500-903, Brazil
| | - Marli Luiza Tebaldi
- Laboratório de Bioengenharia - Universidade Federal de Itajubá (UNIFEI) - Campus Itabira, Itabira, MG 35903-087, Brazil
| |
Collapse
|
23
|
Chowdhury S, Toth I, Stephenson RJ. Dendrimers in vaccine delivery: Recent progress and advances. Biomaterials 2021; 280:121303. [PMID: 34871877 DOI: 10.1016/j.biomaterials.2021.121303] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 11/08/2021] [Accepted: 11/29/2021] [Indexed: 12/19/2022]
Abstract
Dendrimers are well-defined, highly branched, multivalent and monodisperse molecules which host a range of attractive, yet functional, chemical and biological characteristics. A dendrimers accessible surface groups enable coupling to different functional moieties (e.g., antibodies, peptides, proteins, etc), which is further assisted by the dendrimers tailored size and surface charge. This adaptability allows for the preparation of molecularly precise vaccines with highly specific and predictable properties, and in conjunction with a dendrimers immune stimulating (adjuvanting) property, makes dendrimers attractive substrates for biomedical applications, including vaccines. This review highlights the structural and synthetic evolution of dendrimers throughout history, detailing the dendrimers role as both an adjuvant and carrier system for vaccine antigens, in addition to reviewing the development of commercially available vaccines for use in humans.
Collapse
Affiliation(s)
- Silvia Chowdhury
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072 Australia.
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia; School of Pharmacy, The University of Queensland, Brisbane, QLD 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072 Australia.
| | - Rachel J Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia.
| |
Collapse
|
24
|
Alotaibi BS, Buabeid M, Ibrahim NA, Kharaba ZJ, Ijaz M, Murtaza G. Recent strategies driving oral biologic administration. Expert Rev Vaccines 2021; 20:1587-1601. [PMID: 34612121 DOI: 10.1080/14760584.2021.1990044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION High patient compliance, noninvasiveness, and self-administration are the leading features of vaccine delivery through the oral route. The implementation of swift mass vaccination campaigns in pandemic outbreaks fascinates the use of oral vaccination. This approach can elicit both mucosal and systemic immune responses to protect against infection at the surface of the mucosa. AREA COVERED As pathogen entry and spread mainly occurs through the gastrointestinal tract (GIT) mucosal surfaces, oral vaccination may protect and limit disease spread. Oral vaccines target various potential mucosal inductive sites in the GIT, such as the oral cavity, gastric area, and small intestine. Orally delivered vaccines having subunit and nucleic acid pass through various GIT-associated risks, such as the biodegradation of biologics and their reduced absorption. This article presents a summarized review of the existing technologies and prospects for oral vaccination. EXPERT OPINION The intestinal mucosa focuses on current approaches, while future strategies target new mucosal sites, i.e. oral cavity and stomach. Recent developments in biologic delivery through the oral route and their potential use in future oral vaccination are mainly considered.
Collapse
Affiliation(s)
- Badriyah Shadid Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, 346, UAE.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Nihal Abdalla Ibrahim
- Department of Clinical Sciences, Ajman University, Ajman, 346, UAE.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Zelal Jaber Kharaba
- Department of Clinical Sciences, College of Pharmacy, Al-Ain University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Munazza Ijaz
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore, 54000, Pakistan
| |
Collapse
|
25
|
Yang Y, Santamaria P. Evolution of nanomedicines for the treatment of autoimmune disease: From vehicles for drug delivery to inducers of bystander immunoregulation. Adv Drug Deliv Rev 2021; 176:113898. [PMID: 34314782 DOI: 10.1016/j.addr.2021.113898] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022]
Abstract
Over the last two decades, the nanomedicine field has witnessed an explosive growth of research on the development of nanoparticle/microparticle (NP/MP)-based compounds for the treatment of autoimmune diseases. Studies have evaluated compounds generated with a broad range of materials with different shapes, sizes, surface chemistries and structures. A number of active pharmaceutical ingredients, including immunosuppressants, cytokines, nucleotides, peptides, proteins and immunomodulators of various types have been encapsulated into or incorporated onto the surface of these compounds, either individually or in combination, and delivered to animal models of autoimmune inflammation via different administration routes. These NP/MP-based compounds can be categorized into four different groups based on their intended mechanisms of action. Here, we review the engineering designs, the pharmacodynamic and therapeutic correlates and the disease specificity of nanomedicines belonging to each of these groups.
Collapse
Affiliation(s)
- Yang Yang
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1 Canada; Department of Biochemistry and Molecular Biology and Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1, Canada.
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1 Canada; Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona 08036, Spain.
| |
Collapse
|
26
|
Chan Y, Ng SW, Singh SK, Gulati M, Gupta G, Chaudhary SK, Hing GB, Collet T, MacLoughlin R, Löbenberg R, Oliver BG, Chellappan DK, Dua K. Revolutionizing polymer-based nanoparticle-linked vaccines for targeting respiratory viruses: A perspective. Life Sci 2021; 280:119744. [PMID: 34174324 PMCID: PMC8223024 DOI: 10.1016/j.lfs.2021.119744] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022]
Abstract
Viral respiratory tract infections have significantly impacted global health as well as socio-economic growth. Respiratory viruses such as the influenza virus, respiratory syncytial virus (RSV), and the recent SARS-CoV-2 infection (COVID-19) typically infect the upper respiratory tract by entry through the respiratory mucosa before reaching the lower respiratory tract, resulting in respiratory disease. Generally, vaccination is the primary method in preventing virus pathogenicity and it has been shown to remarkably reduce the burden of various infectious diseases. Nevertheless, the efficacy of conventional vaccines may be hindered by certain limitations, prompting the need to develop novel vaccine delivery vehicles to immunize against various strains of respiratory viruses and to mitigate the risk of a pandemic. In this review, we provide an insight into how polymer-based nanoparticles can be integrated with the development of vaccines to effectively enhance immune responses for combating viral respiratory tract infections.
Collapse
Affiliation(s)
- Yinghan Chan
- School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sin Wi Ng
- School of Pharmacy, International Medical University (IMU), Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, India
| | - Sushil Kumar Chaudhary
- Faculty of Pharmacy, DIT University, Mussoorie-Diversion Road, Makkawala, Dehradun 248 009, Uttarakhand, India
| | - Goh Bey Hing
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Trudi Collet
- Innovative Medicines Group, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Dangan, H91 HE94 Galway, Ireland; School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Raimar Löbenberg
- University of Alberta, Faculty of Pharmacy and Pharmaceutical Sciences, Edmonton, AB T6G 2N8, Canada
| | - Brian G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| | - Kamal Dua
- University of Alberta, Faculty of Pharmacy and Pharmaceutical Sciences, Edmonton, AB T6G 2N8, Canada; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia.
| |
Collapse
|
27
|
Zhu FD, Hu YJ, Yu L, Zhou XG, Wu JM, Tang Y, Qin DL, Fan QZ, Wu AG. Nanoparticles: A Hope for the Treatment of Inflammation in CNS. Front Pharmacol 2021; 12:683935. [PMID: 34122112 PMCID: PMC8187807 DOI: 10.3389/fphar.2021.683935] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, an inflammatory response within the central nervous system (CNS), is a main hallmark of common neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), among others. The over-activated microglia release pro-inflammatory cytokines, which induces neuronal death and accelerates neurodegeneration. Therefore, inhibition of microglia over-activation and microglia-mediated neuroinflammation has been a promising strategy for the treatment of neurodegenerative diseases. Many drugs have shown promising therapeutic effects on microglia and inflammation. However, the blood–brain barrier (BBB)—a natural barrier preventing brain tissue from contact with harmful plasma components—seriously hinders drug delivery to the microglial cells in CNS. As an emerging useful therapeutic tool in CNS-related diseases, nanoparticles (NPs) have been widely applied in biomedical fields for use in diagnosis, biosensing and drug delivery. Recently, many NPs have been reported to be useful vehicles for anti-inflammatory drugs across the BBB to inhibit the over-activation of microglia and neuroinflammation. Therefore, NPs with good biodegradability and biocompatibility have the potential to be developed as an effective and minimally invasive carrier to help other drugs cross the BBB or as a therapeutic agent for the treatment of neuroinflammation-mediated neurodegenerative diseases. In this review, we summarized various nanoparticles applied in CNS, and their mechanisms and effects in the modulation of inflammation responses in neurodegenerative diseases, providing insights and suggestions for the use of NPs in the treatment of neuroinflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yu-Jiao Hu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Anesthesia, Southwest Medical University, Luzhou, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qing-Ze Fan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
28
|
Guo Z, Kubiatowicz LJ, Fang RH, Zhang L. Nanotoxoids: Biomimetic Nanoparticle Vaccines against Infections. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Luke J. Kubiatowicz
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program and Moores Cancer Center University of California San Diego La Jolla CA 92093 USA
| |
Collapse
|
29
|
Mishra M, Panda M. Reactive oxygen species: the root cause of nanoparticle-induced toxicity in Drosophila melanogaster. Free Radic Res 2021; 55:671-687. [PMID: 33877010 DOI: 10.1080/10715762.2021.1914335] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nanotechnology is a rapidly developing technology in the twenty-first century. Nanomaterials are extensively used in numerous industries including cosmetics, food, medicines, industries, agriculture, etc. Along with its wide application toxicity is also reported from studies of various model organisms including Drosophila. The toxicity reflects cytotoxicity, genotoxicity, and teratogenicity. The current study correlates the toxicity as a consequence of reactive oxygen species (ROS) generated owing to the presence of nanoparticles with the living cell. ROS mainly includes hydroxyl ions, peroxide ions, superoxide anions, singlet oxygen, and hypochlorous acids. An elevated level of ROS can damage the cells by various means. To protect the body from excess ROS, living cells possess a set of antioxidant enzymes which includes peroxidase, glutathione peroxidase, and catalase. If the antioxidant enzymes cannot nullify the elevated ROS level than DNA damage, cell damage, cytotoxicity, apoptosis, and uncontrolled cell regulations occur resulting in abnormal physiological and genotoxic conditions. Herewith, we are reporting various morphological and physiological defects caused after nanoparticle treatment as a function of redox imbalance.
Collapse
Affiliation(s)
- Monalisa Mishra
- Neural Developmental Biology Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Mrutyunjaya Panda
- Neural Developmental Biology Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
30
|
Bekhouche M, Bolon M, Charriaud F, Lamrayah M, Da Costa D, Primard C, Costantini A, Pasdeloup M, Gobert S, Mallein-Gerin F, Verrier B, Ducret M, Farges JC. Development of an antibacterial nanocomposite hydrogel for human dental pulp engineering. J Mater Chem B 2021; 8:8422-8432. [PMID: 32804177 DOI: 10.1039/d0tb00989j] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hydrogel-based regenerative endodontic procedures (REPs) are considered to be very promising therapeutic strategies to reconstruct the dental pulp (DP) tissue in devitalized human teeth. However, the success of the regeneration process is limited by residual bacteria that may persist in the endodontic space after the disinfection step and contaminate the biomaterial. The aim of this work was to develop an innovative fibrin hydrogel incorporating clindamycin (CLIN)-loaded Poly (d,l) Lactic Acid (PLA) nanoparticles (NPs) to provide the hydrogel with antibacterial properties. CLIN-PLA-NPs were synthesized by a surfactant-free nanoprecipitation method and their microphysical properties were assessed by dynamic light scattering, electrophoretic mobility and scanning electron microscopy. Their antimicrobial efficacy was evaluated on Enteroccocus fæcalis by the determination of the minimal inhibitory concentration (MIC) and the minimal biofilm inhibition and eradication concentrations (MBIC and MBEC). Antibacterial properties of the nanocomposite hydrogel were verified by agar diffusion assays. NP distribution into the hydrogel and release from it were evaluated using fluorescent PLA-NPs. NP cytotoxicity was assessed on DP mesenchymal stem cells (DP-MSCs) incorporated into the hydrogel. Type I collagen synthesis was investigated after 7 days of culture by immunohistochemistry. We found that CLIN-PLA-NPs displayed a drug loading of 10 ± 2 μg per mg of PLA polymer and an entrapment efficiency of 43 ± 7%. Antibiotic loading did not affect NP size, polydispersity index and zeta potential. The MIC for Enterococcus fæcalis was 32 μg mL-1. MBIC50 and MBEC50 were 4 and 16 μg mL-1, respectively. CLIN-PLA-NPs appeared homogenously distributed throughout the hydrogel. CLIN-PLA-NP-loaded hydrogels clearly inhibited E. faecalis growth. DP-MSC viability and type I collagen synthesis within the fibrin hydrogel were not affected by CLIN-PLA-NPs. In conclusion, CLIN-PLA-NP incorporation into the fibrin hydrogel gave the latter antibacterial and antibiofilm properties without affecting cell viability and function. This formulation could help establish an aseptic environment supporting DP reconstruction and, accordingly, might be a valuable tool for REPs.
Collapse
Affiliation(s)
- M Bekhouche
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France and Faculté d'Odontologie, Université de Lyon, Université Lyon 1, Lyon, France
| | - M Bolon
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France
| | - F Charriaud
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France
| | - M Lamrayah
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France
| | - D Da Costa
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France and Adjuvatis®, Lyon, France
| | | | - A Costantini
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France
| | - M Pasdeloup
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France
| | - S Gobert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France
| | - F Mallein-Gerin
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France
| | - B Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France
| | - M Ducret
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France and Faculté d'Odontologie, Université de Lyon, Université Lyon 1, Lyon, France and Hospices Civils de Lyon, Service de Consultations et Traitements Dentaires, Lyon, France
| | - J-C Farges
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR5305 CNRS/Université Lyon 1, Lyon, France and Faculté d'Odontologie, Université de Lyon, Université Lyon 1, Lyon, France and Hospices Civils de Lyon, Service de Consultations et Traitements Dentaires, Lyon, France
| |
Collapse
|
31
|
Morelli L, Polito L, Richichi B, Compostella F. Glyconanoparticles as tools to prevent antimicrobial resistance. Glycoconj J 2021; 38:475-490. [PMID: 33728545 PMCID: PMC7964520 DOI: 10.1007/s10719-021-09988-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/24/2021] [Accepted: 02/28/2021] [Indexed: 01/20/2023]
Abstract
The increased phenomenon of antimicrobial resistance and the slow pace of development of new antibiotics are at the base of a global health concern regarding microbial infections. Antibiotic resistance kills an estimated 700,000 people each year worldwide, and this number is expected to increase dramatically if efforts are not made to develop new drugs or alternative containment strategies. Increased vaccination coverage, improved sanitation or sustained implementation of infection control measures are among the possible areas of action. Indeed, vaccination is one of the most effective tools of preventing infections. Starting from 1970s polysaccharide-based vaccines against Meningococcus, Pneumococcus and Haemophilus influenzae type b have been licensed, and provided effective protection for population. However, the development of safe and effective vaccines for infectious diseases with broad coverage remains a major challenge in global public health. In this scenario, nanosystems are receiving attention as alternative delivery systems to improve vaccine efficacy and immunogenicity. In this report, we provide an overview of current applications of glyconanomaterials as alternative platforms in the development of new vaccine candidates. In particular, we will focus on nanoparticle platforms, used to induce the activation of the immune system through the multivalent-displacement of saccharide antigens. ![]()
Collapse
Affiliation(s)
- Laura Morelli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Saldini 50, 20133, Milan, Italy
| | - Laura Polito
- National Research Council, CNR-SCITEC, Via G. Fantoli 16/15, 20138, Milan, Italy
| | - Barbara Richichi
- Department of Chemistry 'Ugo Schiff', University of Florence, Via della Lastruccia 13, 50019, Sesto Fiorentino, FI, Italy
| | - Federica Compostella
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Saldini 50, 20133, Milan, Italy.
| |
Collapse
|
32
|
Wang L, Liu C, Qiao F, Li M, Xin H, Chen N, Wu Y, Liu J. Analysis of the cytotoxic effects, cellular uptake and cellular distribution of paclitaxel-loaded nanoparticles in glioblastoma cells in vitro. Exp Ther Med 2021; 21:292. [PMID: 33717235 PMCID: PMC7885080 DOI: 10.3892/etm.2021.9723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma is the most common and aggressive type of brain tumor. Although treatments for glioblastoma have been improved recently, patients still suffer from local recurrence in addition to poor prognosis. Previous studies have indicated that the efficacy of chemotherapeutic or bioactive agents is severely compromised by the blood-brain barrier and the inherent drug resistance of glioblastoma. The present study developed a delivery system to improve the efficiency of delivering therapeutic agents into glioblastoma cells. The anticancer drug paclitaxel (PTX) was packed into nanoparticles that were composed of amphiphilic poly (γ-glutamic-acid-maleimide-co-L-lactide)-1,2-dipalmitoylsn-glycero-3-phosphoethanolaminecopolymer conjugated with targeting moiety transferrin (Tf). The Tf nanoparticles (Tf-NPs) may enter glioblastoma cells via transferrin receptor-mediated endocytosis. MTT assay and flow cytometry were used to explore the cytotoxic effects, cellular uptake and cellular distribution of paclitaxel-loaded nanoparticles. The results indicated that both PTX and PTX-Tf-NPs inhibited the viability of rat glioblastoma C6 cells in a dose-dependent manner, but the PTX-Tf-NPs exhibited a greater inhibitory effect compared with PTX, even at higher concentrations (0.4, 2 and 10 µg/ml). However, both PTX and PTX-Tf-NPs exhibited a reduced inhibitory effect on the viability of mouse hippocampal neuronal HT22 cells compared with that on C6 cells. Additionally, in contrast to PTX alone, PTX-Tf-NPs treatment of C6 cells at lower concentrations (0.0032, 0.0160 and 0.0800 µg/ml) induced increased G2/M arrest, although this difference did not occur at a higher drug concentration (0.4 µg/ml). It was observed that FITC-labeled PTX-Tf-NPs were endocytosed by C6 cells within 4 h. Furthermore, FITC-labeled PTX-Tf-NPs or Tf-NPs co-localized with a lysosomal tracker, Lysotracker Red DND-99. These results of the present study indicated that Tf-NPs enhanced the cytotoxicity of PTX in glioblastoma C6 cells, suggesting that PTX-Tf-NPs should be further explored in animal models of glioblastoma.
Collapse
Affiliation(s)
- Lin Wang
- Clinical Laboratory, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Chunhui Liu
- Clinical Laboratory, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Feng Qiao
- Clinical Laboratory, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Mingjun Li
- Clinical Laboratory, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Hua Xin
- Clinical Laboratory, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Naifeng Chen
- Department of Pathology and Physiology, School of Basic Medical Sciences of Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Yan Wu
- Division of Nanomedicine and Nanobiology, National Center for Nanoscience and Technology, Beijing 100190, P.R. China
| | - Junxing Liu
- Department of Pathology and Physiology, School of Basic Medical Sciences of Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| |
Collapse
|
33
|
Chen D, Liu Y, Zhang Z, Liu Z, Fang X, He S, Wu C. NIR-II Fluorescence Imaging Reveals Bone Marrow Retention of Small Polymer Nanoparticles. NANO LETTERS 2021; 21:798-805. [PMID: 33346668 DOI: 10.1021/acs.nanolett.0c04543] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The concept that systemically administered nanoparticles are highly accumulated into the liver, spleen and kidney is a central paradigm in the field of nanomedicine. Here, we report that bone is an important organ for retention of small polymer nanoparticles using in vivo fluorescence imaging in the second near-infrared (NIR-II) window. We prepared different sized polymer nanoparticles with both visible and NIR-II fluorescence. NIR-II imaging reveals that the behavior of nanoparticle distribution in bone was largely dependent on the particle size. Small polymer nanoparticles of ∼15 nm diameter showed fast accumulation and long-term retention in bone, while the nanoparticles larger than ∼25 nm were dominantly distributed in liver. Confocal microscopy of bone sections indicated that the nanoparticles were largely distributed in the endothelial cells of sinusoidal vessels in bone marrow. The study provides promising opportunities for bone imaging and treatment of bone-related disease.
Collapse
Affiliation(s)
- Dandan Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ye Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhe Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhihe Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xiaofeng Fang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Shuqing He
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Changfeng Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
34
|
Coffey JW, Gaiha GD, Traverso G. Oral Biologic Delivery: Advances Toward Oral Subunit, DNA, and mRNA Vaccines and the Potential for Mass Vaccination During Pandemics. Annu Rev Pharmacol Toxicol 2021; 61:517-540. [PMID: 32466690 PMCID: PMC8057107 DOI: 10.1146/annurev-pharmtox-030320-092348] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Oral vaccination enables pain-free and self-administrable vaccine delivery for rapid mass vaccination during pandemic outbreaks. Furthermore, it elicits systemic and mucosal immune responses. This protects against infection at mucosal surfaces, which may further enhance protection and minimize the spread of disease. The gastrointestinal (GI) tract presents a number of prospective mucosal inductive sites for vaccine targeting, including the oral cavity, stomach, and small intestine. However, currently available oral vaccines are effectively limited to live-attenuated and inactivated vaccines against enteric diseases. The GI tract poses a number of challenges,including degradative processes that digest biologics and mucosal barriers that limit their absorption. This review summarizes the approaches currently under development and future opportunities for oral vaccine delivery to established (intestinal) and relatively new (oral cavity, stomach) mucosal targets. Special consideration is given to recent advances in oral biologic delivery that offer promise as future platforms for the administration of oral vaccines.
Collapse
Affiliation(s)
- Jacob William Coffey
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunology, University of Melbourne, Victoria, 3000, Australia
| | - Gaurav Das Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139, USA
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| |
Collapse
|
35
|
Nanoparticles as Adjuvants and Nanodelivery Systems for mRNA-Based Vaccines. Pharmaceutics 2020; 13:pharmaceutics13010045. [PMID: 33396817 PMCID: PMC7823281 DOI: 10.3390/pharmaceutics13010045] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
Messenger RNA (mRNA)-based vaccines have shown promise against infectious diseases and several types of cancer in the last two decades. Their promise can be attributed to their safety profiles, high potency, and ability to be rapidly and affordably manufactured. Now, many RNA-based vaccines are being evaluated in clinical trials as prophylactic and therapeutic vaccines. However, until recently, their development has been limited by their instability and inefficient in vivo transfection. The nanodelivery system plays a dual function in RNA-based vaccination by acting as a carrier system and as an adjuvant. That is due to its similarity to microorganisms structurally and size-wise; the nanodelivery system can augment the response by the immune system via simulating the natural infection process. Nanodelivery systems allow non-invasive mucosal administration, targeted immune cell delivery, and controlled delivery, reducing the need for multiple administrations. They also allow co-encapsulating with immunostimulators to improve the overall adjuvant capacity. The aim of this review is to discuss the recent developments and applications of biodegradable nanodelivery systems that improve RNA-based vaccine delivery and enhance the immunological response against targeted diseases.
Collapse
|
36
|
Carmona-Ribeiro AM, Pérez-Betancourt Y. Cationic Nanostructures for Vaccines Design. Biomimetics (Basel) 2020; 5:biomimetics5030032. [PMID: 32645946 PMCID: PMC7560170 DOI: 10.3390/biomimetics5030032] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
Subunit vaccines rely on adjuvants carrying one or a few molecular antigens from the pathogen in order to guarantee an improved immune response. However, to be effective, the vaccine formulation usually consists of several components: an antigen carrier, the antigen, a stimulator of cellular immunity such as a Toll-like Receptors (TLRs) ligand, and a stimulator of humoral response such as an inflammasome activator. Most antigens are negatively charged and combine well with oppositely charged adjuvants. This explains the paramount importance of studying a variety of cationic supramolecular assemblies aiming at the optimal activity in vivo associated with adjuvant simplicity, positive charge, nanometric size, and colloidal stability. In this review, we discuss the use of several antigen/adjuvant cationic combinations. The discussion involves antigen assembled to 1) cationic lipids, 2) cationic polymers, 3) cationic lipid/polymer nanostructures, and 4) cationic polymer/biocompatible polymer nanostructures. Some of these cationic assemblies revealed good yet poorly explored perspectives as general adjuvants for vaccine design.
Collapse
|
37
|
Meena J, Kumar R, Singh M, Ahmed A, Panda AK. Modulation of immune response and enhanced clearance of Salmonella typhi by delivery of Vi polysaccharide conjugate using PLA nanoparticles. Eur J Pharm Biopharm 2020; 152:270-281. [DOI: 10.1016/j.ejpb.2020.05.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/17/2020] [Accepted: 05/24/2020] [Indexed: 10/24/2022]
|
38
|
Schmidt SJ, Holt BD, Arnold AM, Sydlik SA. Polyester functional graphenic materials as a mechanically enhanced scaffold for tissue regeneration. RSC Adv 2020; 10:8548-8557. [PMID: 35497868 PMCID: PMC9049999 DOI: 10.1039/c9ra10646d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/19/2020] [Indexed: 12/31/2022] Open
Abstract
Traditional metal implants such as titanium, cobalt, and chromium have found wide utility in medicine; however, these come with a risk of toxicity. To overcome metal-related toxicity and enable degradability, polyesters including polycaprolactone (PCL), polylactic acid (PLA), and polyglycolic acid (PGA) show promise for the replacement of various biomedical applications of metals due to their accepted biocompatibility and FDA approval. However, polyesters are less stiff than their metallic counterparts, limiting their application to non-load bearing injury sites, such as fixation hardware for fingers. To improve mechanical properties, graphene oxide (GO)-polyester composites are a promising class of biodegradable scaffolds. Initial reports of these composites are encouraging, but mechanical properties still fall short. Traditional composites rely on non-covalent association between GO and the polyesters, which often leads to failure at the interface and weakens the overall strength of the material. Herein, we present a strategy for attachment of these FDA-approved polyesters onto a derivative of GO using a robust covalent bond. By covalently functionalizing the graphenic backbone with polyesters and without metal catalysts, we create functional graphenic materials (FGMs) to not only simultaneously retain biodegradability and compatibility, but also mechanically strengthen PCL, PLA, and PGA; we observed an average increase in the Young's modulus of over 140% compared to the graphenic backbone. These polyester-functionalized FGMs are a promising platform technology for tissue implants.
Collapse
Affiliation(s)
| | - Brian D Holt
- Carnegie Mellon University 4400 Fifth Ave Pittsburgh PA 15213 USA
| | - Anne M Arnold
- Carnegie Mellon University 4400 Fifth Ave Pittsburgh PA 15213 USA
| | | |
Collapse
|
39
|
Vacchini M, Edwards R, Guizzardi R, Palmioli A, Ciaramelli C, Paiotta A, Airoldi C, La Ferla B, Cipolla L. Glycan Carriers As Glycotools for Medicinal Chemistry Applications. Curr Med Chem 2019; 26:6349-6398. [DOI: 10.2174/0929867326666190104164653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 11/07/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022]
Abstract
Carbohydrates are one of the most powerful and versatile classes of biomolecules that nature
uses to regulate organisms’ biochemistry, modulating plenty of signaling events within cells, triggering
a plethora of physiological and pathological cellular behaviors. In this framework, glycan carrier
systems or carbohydrate-decorated materials constitute interesting and relevant tools for medicinal
chemistry applications. In the last few decades, efforts have been focused, among others, on the development
of multivalent glycoconjugates, biosensors, glycoarrays, carbohydrate-decorated biomaterials
for regenerative medicine, and glyconanoparticles. This review aims to provide the reader with a general
overview of the different carbohydrate carrier systems that have been developed as tools in different
medicinal chemistry approaches relying on carbohydrate-protein interactions. Given the extent of
this topic, the present review will focus on selected examples that highlight the advancements and potentialities
offered by this specific area of research, rather than being an exhaustive literature survey of
any specific glyco-functionalized system.
Collapse
Affiliation(s)
- Mattia Vacchini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Rana Edwards
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Roberto Guizzardi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Alessandro Palmioli
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Carlotta Ciaramelli
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Alice Paiotta
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Barbara La Ferla
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Laura Cipolla
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| |
Collapse
|
40
|
Jawinski K, Hartmann M, Singh C, Kinnear E, Busse DC, Ciabattini A, Fiorino F, Medaglini D, Trombetta CM, Montomoli E, Contreras V, Le Grand R, Coiffier C, Primard C, Verrier B, Tregoning JS. Recombinant Haemagglutinin Derived From the Ciliated Protozoan Tetrahymena thermophila Is Protective Against Influenza Infection. Front Immunol 2019; 10:2661. [PMID: 31798589 PMCID: PMC6863932 DOI: 10.3389/fimmu.2019.02661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
Current influenza vaccines manufactured using eggs have considerable limitations, both in terms of scale up production and the potential impact passaging through eggs can have on the antigenicity of the vaccine virus strains. Alternative methods of manufacture are required, particularly in the context of an emerging pandemic strain. Here we explore the production of recombinant influenza haemagglutinin using the ciliated protozoan Tetrahymena thermophila. For the first time we were able to produce haemagglutinin from both seasonal influenza A and B strains. This ciliate derived material was immunogenic, inducing an antibody response in both mice and non-human primates. Mice immunized with ciliate derived haemagglutinin were protected against challenge with homologous influenza A or B viruses. The antigen could also be combined with submicron particles containing a Nod2 ligand, significantly boosting the immune response and reducing the dose of antigen required. Thus, we show that Tetrahymena can be used as a manufacturing platform for viral vaccine antigens.
Collapse
Affiliation(s)
| | | | - Charanjit Singh
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, United Kingdom
| | - Ekaterina Kinnear
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, United Kingdom
| | - David C Busse
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, United Kingdom
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.,VisMederi s.r.l., Siena, Italy
| | - Vanessa Contreras
- CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Le Kremlin-Bicêtre, France
| | - Roger Le Grand
- CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Le Kremlin-Bicêtre, France
| | - Celine Coiffier
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France
| | | | | | - John S Tregoning
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, United Kingdom
| |
Collapse
|
41
|
Jin Z, Gao S, Cui X, Sun D, Zhao K. Adjuvants and delivery systems based on polymeric nanoparticles for mucosal vaccines. Int J Pharm 2019; 572:118731. [PMID: 31669213 DOI: 10.1016/j.ijpharm.2019.118731] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 09/22/2019] [Accepted: 09/23/2019] [Indexed: 02/07/2023]
Abstract
Most pathogens enter the body through mucosal surfaces. Therefore, vaccination through the mucosal route can greatly enhance the mucosal immune response. Vaccination via the mucosal surface is the most effective way to trigger a protective mucosal immune response, but the vast majority of vaccines used are administered by injection. Strategies to enhance the mucosal immunity have been developed by using vaccine adjuvants, delivery systems, bacterial or viral vectors, and DNA vaccines. Appropriate vaccine adjuvants and drug delivery systems can improve the immunogenicity of antigens, induce a stronger immune response, and reduce the vaccine dose and production cost. In recent years, many studies have focused on finding safe and effective vaccine adjuvants and drug delivery systems to formulate the mucosal vaccines for solving the above problems. Great progress has also been made in vaccine adjuvants and drug delivery systems based on biodegradable polymer nanoparticles. In this paper, the research progress of the mucosal vaccine and its related adjuvants and drug delivery systems in recent years was reviewed, and the application of polymers as adjuvants and drug delivery system in vaccine was prospected. This review provides a fundamental knowledge for the application of biodegradable polymer nanoparticles as adjuvants and carriers in mucosal vaccines and shows great application prospects.
Collapse
Affiliation(s)
- Zheng Jin
- Key Laboratory of Chemical Engineering Process and Technology for High-efficiency Conversion, College of Chemistry and Material Sciences, Heilongjiang University, Harbin 150080, China
| | - Shuang Gao
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang University, Harbin 150080, China; Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin 150080, China
| | - Xianlan Cui
- Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin 150080, China; Bluesky Biotech (Harbin) Co., Ltd., Harbin 150028, China
| | - Dejun Sun
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang University, Harbin 150080, China.
| | - Kai Zhao
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang University, Harbin 150080, China; Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin 150080, China.
| |
Collapse
|
42
|
Tulbah AS, Pisano E, Landh E, Scalia S, Young PM, Traini D, Ong HX. Simvastatin Nanoparticles Reduce Inflammation in LPS-Stimulated Alveolar Macrophages. J Pharm Sci 2019; 108:3890-3897. [PMID: 31494116 DOI: 10.1016/j.xphs.2019.08.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
Abstract
Simvastatin (SV) is widely used as a lipid-lowering medication that has also been found to have beneficial immunomodulatory effects for treatment of chronic lung diseases. Although its anti-inflammatory activity has been investigated, its underlying mechanisms have not yet been clearly elucidated. In this study, the anti-inflammatory and antioxidant effects and mechanism of simvastatin nanoparticles (SV-NPs) on lipopolysaccharide-stimulated alveolar macrophages (AMs) NR8383 cells were investigated. Quantitative cellular uptake of SV-NPs, the production of inflammatory mediators (interleukin-6, tumor necrosis factor, and monocyte chemoattractant protein-1), and oxidative stress (nitric oxide) were tested. Furthermore, the involvement of the nuclear factor κB (NF-κB) signaling pathway in activation of inflammation in AMs and the efficacy of SV were visualized using immunofluorescence. Results indicated that SV-NPs exhibit a potent inhibitory effect on nitric oxide production and secretion of inflammatory cytokine in inflamed AM, without affecting cell viability. The enhanced anti-inflammatory activity of SV-NPs is likely due to SV-improved chemical-physical stability and higher cellular uptake into AM. The study also indicates that SV targets the inflammatory and oxidative response of AM, through inactivation of the NF-κB signaling pathway, supporting the pharmacological basis of SV for treatment of chronic inflammatory lung diseases.
Collapse
Affiliation(s)
- Alaa S Tulbah
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia; College of Pharmacy, Umm Al Qura University, Makkah, Saudi Arabia
| | - Elvira Pisano
- Dipartimento di Scienze della vita e biotecnologie, University of Ferrara, Italy
| | - Emelie Landh
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia
| | - Santo Scalia
- Dipartimento di Scienze della vita e biotecnologie, University of Ferrara, Italy
| | - Paul M Young
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, 2037 New South Wales, Australia.
| |
Collapse
|
43
|
Saito E, Kuo R, Kramer KR, Gohel N, Giles DA, Moore BB, Miller SD, Shea LD. Design of biodegradable nanoparticles to modulate phenotypes of antigen-presenting cells for antigen-specific treatment of autoimmune disease. Biomaterials 2019; 222:119432. [PMID: 31480002 DOI: 10.1016/j.biomaterials.2019.119432] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/18/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Current therapeutic options for autoimmune diseases, such as multiple sclerosis (MS), often require lifelong treatment with immunosuppressive drugs, yet strategies for antigen-specific immunomodulation are emerging. Biodegradable particles loaded with disease-specific antigen, either alone or with immunomodulators, have been reported to ameliorate disease. Herein, we hypothesized that the carrier could impact polarization of the immune cells that associate with particles and the subsequent disease progression. Single injection of three polymeric carriers, 50:50 poly (DL-lactide-co-glycolide) (PLG) with two molecular weights (Low, High) and poly (DL-lactide) (PLA), loaded with the disease-specific antigen, proteolipid protein (PLP139-151), were investigated for the ability to attenuate clinical scores in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. At a low particle dose, mice treated with PLA-based particles had significantly lower clinical scores at the chronic stage of the disease over 200 days post immunization, while neither PLG-based particles nor OVA control particles reduced the clinical scores. Compared to PLG-based particles, PLA-based particles were largely associated with Kupffer cells and liver sinusoidal endothelial cells, which had a reduced co-stimulatory molecule expression that correlated with a reduction of CD4+ T-cell populations in the central nervous system. Delivery of PLA-based particles encapsulated with higher levels of PLP139-151 at a reduced dose were able to completely ameliorate EAE over 200 days along with inhibition of Th1 and Th17 polarization. Collectively, our study demonstrates that the carrier properties and antigen loading determine phenotypes of immune cells in the peripheral organs, influencing the amelioration of both acute and chronic stages of autoimmunity.
Collapse
Affiliation(s)
- Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert Kuo
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kevin R Kramer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nishant Gohel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David A Giles
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Bethany B Moore
- Department of Immunology, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, 60208, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, 60611, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
44
|
Song X, Wang J, Xu Y, Shao H, Gu J. Surface-modified PLGA nanoparticles with PEG/LA-chitosan for targeted delivery of arsenic trioxide for liver cancer treatment: Inhibition effects enhanced and side effects reduced. Colloids Surf B Biointerfaces 2019; 180:110-117. [PMID: 31030022 DOI: 10.1016/j.colsurfb.2019.04.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/26/2019] [Accepted: 04/15/2019] [Indexed: 11/17/2022]
Abstract
Arsenic trioxide (As2O3), an effective drug for leukemia, is limited to be used for solid tumor treatment due to its high side effects. In this study, polyethylene glycol (PEG) and lactobionic acid (LA) modified chitosan (PLC) was synthesized and was used to coat poly(lactide-co-glycolide) (PLGA) nanoparticles for encapsulation and targeted release of As2O3 in liver cancer treatment. The As2O3-loaded PLGA/PLC nanoparticles (As2O3-PLGA/PLC NPs) were fabricated through double emulsion-solvent evaporation method and were optimized by orthogonal tests. As2O3-PLGA/PLC NPs presented suitable physical stability, positive charge, high encapsulation efficiency and drug loading, and good biocompatibility. As expected, the NPs can quickly release enough dose of As2O3 in a short time and then sustain the drug concentration. The As2O3-PLGA/PLC NPs showed effective inhibition of SMMC-7721 cells while having lower cytotoxicity against normal human liver cells (LO2 cells). Furthermore, In vivo study showed that the NPs did not present toxic effects on kidney and liver, but showed relatively high growth inhibition effect on liver tumor. Therefore, this PLGA/PLC NPs could be an effective and safe drug delivery system for liver cancer chemotherapy.
Collapse
Affiliation(s)
- Xiaoli Song
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, PR China.
| | - Juan Wang
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, PR China
| | - Yue Xu
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225002, PR China
| | - Hongxia Shao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225002, PR China
| | - Jun Gu
- Xishan People's Hospital, Wuxi, 214011, PR China.
| |
Collapse
|
45
|
Koerner J, Horvath D, Groettrup M. Harnessing Dendritic Cells for Poly (D,L-lactide- co-glycolide) Microspheres (PLGA MS)-Mediated Anti-tumor Therapy. Front Immunol 2019; 10:707. [PMID: 31024545 PMCID: PMC6460768 DOI: 10.3389/fimmu.2019.00707] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
With emerging success in fighting off cancer, chronic infections, and autoimmune diseases, immunotherapy has become a promising therapeutic approach compared to conventional therapies such as surgery, chemotherapy, radiation therapy, or immunosuppressive medication. Despite the advancement of monoclonal antibody therapy against immune checkpoints, the development of safe and efficient cancer vaccine formulations still remains a pressing medical need. Anti-tumor immunotherapy requires the induction of antigen-specific CD8+ cytotoxic T lymphocyte (CTL) responses which recognize and specifically destroy tumor cells. Due to the crucial role of dendritic cells (DCs) in initiating anti-tumor immunity, targeting tumor antigens to DCs has become auspicious in modern vaccine research. Over the last two decades, micron- or nanometer-sized particulate delivery systems encapsulating tumor antigens and immunostimulatory molecules into biodegradable polymers have shown great promise for the induction of potent, specific and long-lasting anti-tumor responses in vivo. Enhanced vaccine efficiency of the polymeric micro/nanoparticles has been attributed to controlled and continuous release of encapsulated antigens, efficient targeting of antigen presenting cells (APCs) such as DCs and subsequent induction of CTL immunity. Poly (D, L-lactide-co-glycolide) (PLGA), as one of these polymers, has been extensively studied for the design and development of particulate antigen delivery systems in cancer therapy. This review provides an overview of the current state of research on the application of PLGA microspheres (PLGA MS) as anti-tumor cancer vaccines in activating and potentiating immune responses attempting to highlight their potential in the development of cancer therapeutics.
Collapse
Affiliation(s)
- Julia Koerner
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Dennis Horvath
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Marcus Groettrup
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany.,Biotechnology Institute Thurgau at the University of Konstanz, Kreuzlingen, Switzerland
| |
Collapse
|
46
|
Tsatsakis A, Stratidakis AK, Goryachaya AV, Tzatzarakis MN, Stivaktakis PD, Docea AO, Berdiaki A, Nikitovic D, Velonia K, Shtilman MI, Rizos AK, Kuskov AN. In vitro blood compatibility and in vitro cytotoxicity of amphiphilic poly-N-vinylpyrrolidone nanoparticles. Food Chem Toxicol 2019; 127:42-52. [PMID: 30836108 DOI: 10.1016/j.fct.2019.02.041] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 12/11/2022]
Abstract
This study focused on defining the in vitro behavior of amphiphilic poly-N-vinylpyrrolidone (Amph-PVP) nanoparticles toward whole blood, blood plasma and blood cells in order to assess nanoparticle blood compatibility. In addition, possible effects on endothelium cell growth/viability were evaluated. The Amph-PVP nanoparticles were formed via self-assembling in aqueous media and composed of a hydrophobic alkyl core and a hydrophilic PVP outer shell. Their blood compatibility was evaluated by investigating their effect on red blood cells (RBCs) or erythrocytes, white blood cells (WBCs) or leukocytes, platelets (PLTs) and on complement system activation. Our results clearly demonstrate that the Amph-PVP nanoparticles are stable in presence of blood serum, have no significant effects on the function of RBCs, WBCs, PLTs and complement system activation. The Amph-PVP nanoparticles did not show considerable hemolytic or inflammatory effect, neither influence on platelet aggregation, coagulation process, or complement activation at the tested concentration range of 0.05-0.5 mg/ml. The Amph-PVP nanoparticles did not exhibit any significant effect on HMEC-1 microvascular skin endothelial cells' growth in in vitro experiments. The excellent blood compatibility of the Amph-PVP nanoparticles and the lack of effect on endothelium cell growth/viability represent a crucial feature dictating their further study as novel drug delivery systems.
Collapse
Affiliation(s)
- A Tsatsakis
- Laboratory of Toxicology, University of Crete, Voutes, Heraklion, 71003, Crete, Greece; Department of Biomaterials, D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation; Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation.
| | - A K Stratidakis
- Laboratory of Toxicology, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - A V Goryachaya
- Department of Biomaterials, D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation
| | - M N Tzatzarakis
- Laboratory of Toxicology, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - P D Stivaktakis
- Laboratory of Toxicology, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - A O Docea
- Department of Toxicology, University of Medicine and Pharmacy, Faculty of Pharmacy, Craiova, Romania
| | - Ai Berdiaki
- Laboratory of Anatomy-Histology-Embryology, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - D Nikitovic
- Laboratory of Anatomy-Histology-Embryology, University of Crete, Voutes, Heraklion, 71003, Crete, Greece
| | - K Velonia
- Department of Materials Science and Technology, University of Crete, University Campus Voutes, Heraklion, 71003, Crete, Greece
| | - M I Shtilman
- Department of Biomaterials, D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation
| | - A K Rizos
- Department of Chemistry, University of Crete, Foundation for Research and Technology-Hellas, FORTH-IESL, Heraklion, 71003, Crete, Greece
| | - A N Kuskov
- Department of Biomaterials, D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation; Department of Technology of Chemical Pharmaceutical and Cosmetic Products, D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russian Federation.
| |
Collapse
|
47
|
Jeon SG, Cha MY, Kim JI, Hwang TW, Kim KA, Kim TH, Song KC, Kim JJ, Moon M. Vitamin D-binding protein-loaded PLGA nanoparticles suppress Alzheimer's disease-related pathology in 5XFAD mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 17:297-307. [PMID: 30794963 DOI: 10.1016/j.nano.2019.02.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 01/23/2019] [Accepted: 02/07/2019] [Indexed: 10/27/2022]
Abstract
The aggregation and accumulation of amyloid beta (Aβ) peptide is believed to be the primary cause of Alzheimer's disease (AD) pathogenesis. Vitamin D-binding protein (DBP) can attenuate Aβ aggregation and accumulation. A biocompatible polymer poly (D,L-lactic acid-co-glycolic acid) (PLGA) can be loaded with therapeutic agents and control the rate of their release. In the present study, a PLGA-based drug delivery system was used to examine the therapeutic effects of DBP-PLGA nanoparticles in Aβ-overexpressing (5XFAD) mice. DBP was loaded into PLGA nanoparticles and the characteristics of the DBP-PLGA nanoparticles were analyzed. Using a thioflavin-T assay, we observed that DBP-PLGA nanoparticles significantly inhibited Aβ aggregation in vitro. In addition, we found that intravenous injection of DBP-PLGA nanoparticles significantly attenuated the Aβ accumulation, neuroinflammation, neuronal loss and cognitive dysfunction in the 5XFAD mice. Collectively, our results suggest that DBP-PLGA nanoparticles could be a promising therapeutic candidate for the treatment of AD.
Collapse
Affiliation(s)
- Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Moon-Yong Cha
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jin-Il Kim
- Department of Nursing, College of Nursing, Jeju National University, Jeju-si, Republic of Korea
| | - Tae Woong Hwang
- Department of Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kyoung Ah Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Tae Hyoung Kim
- Department of Biomedical Materials, Konyang University, Daejeon, Republic of Korea
| | - Ki Chang Song
- Department of Biomedical Materials, Konyang University, Daejeon, Republic of Korea
| | - Jwa-Jin Kim
- Department of Biomedical Science, Jungwon University, Goesan-gun, Chungbuk, Republic of Korea; Department of Nephrology, School of Medicine, Chungnam National University, Daejeon, Republic of Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea.
| |
Collapse
|
48
|
Coolen AL, Lacroix C, Mercier-Gouy P, Delaune E, Monge C, Exposito JY, Verrier B. Poly(lactic acid) nanoparticles and cell-penetrating peptide potentiate mRNA-based vaccine expression in dendritic cells triggering their activation. Biomaterials 2018; 195:23-37. [PMID: 30610991 DOI: 10.1016/j.biomaterials.2018.12.019] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023]
Abstract
Messenger RNA-based vaccines have the potential to trigger robust cytotoxic immune responses, which are essential for fighting cancer and infectious diseases like HIV. Dendritic Cells (DCs) are choice targets for mRNA-based vaccine strategies, as they link innate and adaptive immune responses and are major regulators of cytotoxic and humoral adaptive responses. However, efficient delivery of antigen-coding mRNAs into DC cytosol has been highly challenging. In this study, we developed an alternative to lipid-based mRNA delivery systems, using poly(lactic acid) nanoparticles (PLA-NPs) and cationic cell-penetrating peptides as mRNA condensing agent. The formulations are assembled in two steps: (1) formation of a polyplex between mRNAs and amphipathic cationic peptides (RALA, LAH4 or LAH4-L1), and (2) adsorption of polyplexes onto PLA-NPs. LAH4-L1/mRNA polyplexes and PLA-NP/LAH4-L1/mRNA nanocomplexes are taken up by DCs via phagocytosis and clathrin-dependent endocytosis, and induce strong protein expression in DCs in vitro. They modulate DC innate immune response by activating both endosome and cytosolic Pattern Recognition Receptors (PRRs), and induce markers of adaptive responses in primary human DCs in vitro, with prevalent Th1 signature. Thus, LAH4-L1/mRNA and PLA-NP/LAH4-L1/mRNA represent a promising platform for ex vivo treatment and mRNA vaccine development.
Collapse
Affiliation(s)
- Anne-Line Coolen
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France
| | - Céline Lacroix
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France
| | - Perrine Mercier-Gouy
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France
| | - Emilie Delaune
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France
| | - Claire Monge
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France
| | - Jean-Yves Exposito
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France.
| |
Collapse
|
49
|
Ferrari R, Sponchioni M, Morbidelli M, Moscatelli D. Polymer nanoparticles for the intravenous delivery of anticancer drugs: the checkpoints on the road from the synthesis to clinical translation. NANOSCALE 2018; 10:22701-22719. [PMID: 30512025 DOI: 10.1039/c8nr05933k] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
In this review article we discuss some of the key aspects concerning the development of a polymer-based nanoparticle formulation for intravenous drug delivery. Since numerous preparations fail before and during clinical trials, our aim is to emphasize the main issues that a nanocarrier has to face once injected into the body. These include biocompatibility and toxicity, drug loading and release, nanoparticle storage and stability, biodistribution, selectivity towards the target organs or tissues, internalization in cells and biodegradability. They represent the main checkpoints to define a polymer-based formulation as safe and effective. Indeed, this review is intended to provide guidelines to be followed in the early development of a new nanotherapeutic to hopefully increase the success rate of polymer-based formulations entering clinical trials. The corresponding requirements and characteristics are discussed in the context of some relevant case studies taken from the literature and mainly related to the delivery of lipophilic anticancer therapeutics.
Collapse
Affiliation(s)
- R Ferrari
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1, 8093, Zurich, Switzerland.
| | - M Sponchioni
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1, 8093, Zurich, Switzerland. and Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy
| | - M Morbidelli
- Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1, 8093, Zurich, Switzerland.
| | - D Moscatelli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy
| |
Collapse
|
50
|
Song X, You J, Shao H, Yan C. Effects of surface modification of As 2 O 3 -loaded PLGA nanoparticles on its anti-liver cancer ability: An in vitro and in vivo study. Colloids Surf B Biointerfaces 2018; 169:289-297. [DOI: 10.1016/j.colsurfb.2018.05.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 04/16/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023]
|