1
|
Hu H, Chen WJ, Sun C, Xie JP. Progress in the development of cancer vaccines for lung cancer utilizing dendritic cells (Review). Oncol Lett 2025; 29:298. [PMID: 40276084 PMCID: PMC12018796 DOI: 10.3892/ol.2025.15044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/20/2024] [Indexed: 04/26/2025] Open
Abstract
Lung cancer is a major global health concern in terms of both incidence and mortality. Despite substantial advancements in targeted therapy and immune checkpoint inhibitor treatments, their overall effectiveness is limited. Dendritic cells (DCs) are crucial in innate and acquired immune responses due to their effective presentation of antigens. DC-based cancer vaccines have been identified as promising strategies for personalized cancer immunotherapy. The present review presents a thorough examination of the immunomodulation and associated mechanisms of DC vaccines in lung cancer, with a specific emphasis on the presentation of clinical trial data concerning the safety, feasibility and effectiveness of DC vaccines in the treatment of patients with lung cancer. The objective of this review is to highlight strategies and provide insights that may improve the development and clinical efficacy of DC vaccines for the treatment lung cancer.
Collapse
Affiliation(s)
- Hui Hu
- Department of Respiratory and Critical Care, The Second Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wen-Jun Chen
- Department of Oncology, PLA Navy Anqing Hospital, Anqing, Anhui 246004, P.R. China
| | - Chuang Sun
- Department of Respiratory and Critical Care, Xinyu People's Hospital, Xinyu, Jiangxi 338099, P.R. China
| | - Jun-Ping Xie
- Department of Respiratory and Critical Care, The Second Affiliated Hospital of Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
2
|
Li Z, Chen P, Qu A, Sun M, Xu L, Xu C, Hu S, Kuang H. Opportunities and Challenges for Nanomaterials as Vaccine Adjuvants. SMALL METHODS 2025:e2402059. [PMID: 40277301 DOI: 10.1002/smtd.202402059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/29/2025] [Indexed: 04/26/2025]
Abstract
Adjuvants, as a critical component of vaccines, are capable of eliciting more robust and sustained immune responses. Nanomaterials have shown unique advantages and broad application prospects in adjuvant development due to their high adjustability and distinctive physicochemical properties. This review focuses on nanoadjuvants and their immunological mechanisms. First, various types of adjuvants are introduced with an emphasis on metal and metal oxide nanoparticles, coordination polymers, liposomes, polymer nanoparticles, and other inorganic nanoparticles that can serve as vaccine adjuvants. Second, this review describes the current status of the clinical applications of nanoadjuvants. Next, the mechanisms of action for nanoadjuvants have been thoroughly elucidated, including the depot effect, NLRP3 inflammasome activation, targeting C-type lectin receptors, activation of toll-like receptors, and activation of the cGAS-STING signaling pathway. Finally, the challenges and opportunities associated with the development of nanoadjuvants have also been addressed.
Collapse
Affiliation(s)
- Zongda Li
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Panpan Chen
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aihua Qu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Shudong Hu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
3
|
Vijayanand S, Patil S, Bagwe P, Singh R, Adediran E, D’Souza MJ. Evaluating the Immunogenicity of an Intranasal Microparticle Combination Vaccine for COVID-19 and Influenza. Vaccines (Basel) 2025; 13:282. [PMID: 40266139 PMCID: PMC11946802 DOI: 10.3390/vaccines13030282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Infectious respiratory pathogens like SARS-CoV-2 and influenza frequently mutate, leading to the emergence of variants. This necessitates continuous updates to FDA-approved vaccines with booster shots targeting the circulating variants. Vaccine hesitancy and needle injections create inconvenience and contribute to reduced global vaccination rates. To address the burden of frequent painful injections, this manuscript explores the potential of non-invasive intranasal (IN) vaccine administration as an effective alternative to intramuscular (IM) shots. Further, as a proof-of-concept, an inactivated combination vaccine for COVID-19 and influenza was tested to eliminate the need for separate vaccinations. METHODS The methods involved encapsulating antigens and adjuvants in poly(lactic-co-glycolic acid) (PLGA) polymer matrices, achieving over 85% entrapment. The vaccine was evaluated in vitro for cytotoxicity and immunogenicity before being administered to 6-8-week-old Swiss Webster mice at weeks 0, 3, and 6. The mice were then assessed for antibody levels and cellular responses. RESULTS The intranasal microparticle (IN-MP) vaccine induced an innate immune response, autophagy, and were non-cytotoxic in vitro. In vivo, the vaccine led to high levels of virus-specific serum IgM, IgG, and IgA binding antibodies, as well as elevated IgG and IgA levels in the lung wash samples. The antibodies generated demonstrated neutralizing activity against the SARS-CoV-2 pseudovirus. Furthermore, the IN-MP vaccine prompted increased antigen-specific CD4+ and CD8+ T-cell responses in the vaccinated mice. CONCLUSIONS The IN-MP combination vaccine produced immune responses comparable to or higher than the IM route, indicating its potential as an alternative to IM injections.
Collapse
Affiliation(s)
| | | | | | | | | | - Martin J. D’Souza
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (S.V.); (S.P.); (P.B.); (R.S.); (E.A.)
| |
Collapse
|
4
|
Amankwa CE, DebNath B, Pham JH, Johnson GA, Zhang W, Ranjan A, Stankowska DL, Acharya S. Optimized PLGA encapsulated SA-2 nanosuspension exhibits sustained intraocular pressure reduction in the mouse microbead occlusion model of ocular hypertension. Eur J Pharm Sci 2025; 206:107016. [PMID: 39827971 DOI: 10.1016/j.ejps.2025.107016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Elevated intraocular pressure (IOP) is implicated in the structural and functional damage to the retinal ganglion cells (RGCs) in primary open-angle glaucoma (POAG). Topical IOP lowering agents provide short-term relief, necessitating frequent dosing. Moreover, non-adherence to frequent eyedrops administration contributes significantly to visual field loss and worsens the disease outcome. We optimized the poly (lactic-co-glycolic acid) (PLGA) nanoparticles encapsulation of hybrid antioxidant-nitric oxide donor SA-2 (SA-2NP), investigated its bioavailability, duration of IOP lowering efficacy, and effects on retinal function in the microbead model of ocular hypertension (OHT). SA-2 was bioavailable in the anterior and posterior segments after 1, 8, and 24 h post-single topical eyedrop administration. SA-2NP significantly lowered IOP (∼25-34%) and preserved the RGC function after weekly eyedrop administration for 3 weeks in C57BL/6J mice. In conclusion, the optimized SA-2NP formulation demonstrated optimal bioavailability, ocular safety, and prolonged IOP-lowering efficacy in the mouse microbead occlusion model of OHT.
Collapse
Affiliation(s)
- Charles E Amankwa
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Biddut DebNath
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Jennifer H Pham
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Gretchen A Johnson
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Wei Zhang
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Amalendu Ranjan
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Dorota L Stankowska
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Suchismita Acharya
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| |
Collapse
|
5
|
Yang Y, Hu Y, Yang Y, Liu Q, Zheng P, Yang Z, Duan B, He J, Li W, Li D, Zheng X, Wang M, Fu Y, Long Q, Ma Y. Tumor Vaccine Exploiting Membranes with Influenza Virus-Induced Immunogenic Cell Death to Decorate Polylactic Coglycolic Acid Nanoparticles. ACS NANO 2025; 19:3115-3134. [PMID: 39806805 DOI: 10.1021/acsnano.4c00654] [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: 01/16/2025]
Abstract
Immunogenic cell death (ICD) of tumor cells, which is characterized by releasing immunostimulatory "find me" and "eat me" signals, expressing proinflammatory cytokines and providing personalized and broad-spectrum tumor antigens draws increasing attention in developing a tumor vaccine. In this study, we aimed to investigate whether the influenza virus (IAV) is efficient enough to induce ICD in tumor cells and an extra modification of IAV components such as hemeagglutinin (HA) will be helpful for the ICD-induced cells to elicit robust antitumor effects; in addition, to evaluate whether the membrane-engineering polylactic coglycolic acid nanoparticles (PLGA NPs) simulating ICD immune stimulation mechanisms hold the potential to be a promising vaccine candidate, a mouse melanoma cell line (B16-F10 cell) was infected with IAV rescued by the reverse genetic system, and the prepared cells and membrane-modified PLGA NPs were used separately to immunize the melanoma-bearing mice. IAV-infected tumor cells exhibit dying status, releasing high mobility group box-1 (HMGB1) and adenosine triphosphate (ATP), and exposing calreticulin (CRT), IAV hemeagglutinin (HA), and tumor antigens like tyrosinase-related protein 2 (TRP2). IAV-induced ICD cells enhance biomass-derived carbon (BMDCs) migration, antigen uptake, cross-presentation, and maturation in vitro. Furthermore, immunization with IAV-induced ICD cells effectively suppressed tumor growth in melanoma-bearing mice. The isolated cell membrane inherited the immunological characteristics from the ICD cells and elicited robust antitumor immune responses through decorating PLGA NPs loading with a tumor-specific helper T-cell peptide and supplemented with ATP in a hydrogel system. This study indicated a promising strategy for developing cell-based and personalized tumor vaccines through fully taking advantage of the immune stimulation mechanisms of ICD occurrence in tumor cells, IAV modification, and nanoscale delivery.
Collapse
Affiliation(s)
- Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Yunnan University, Kunming 650091, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Kunming Medical University, Kunming 650500, China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Center for Disease Control and Prevention, Kunming 650000, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650031, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Kunming 650031, China
| |
Collapse
|
6
|
Gao F, Feng X, Li X. Recent advances in polymeric nanoparticles for the treatment of hepatic diseases. Front Pharmacol 2025; 16:1528752. [PMID: 39925843 PMCID: PMC11802823 DOI: 10.3389/fphar.2025.1528752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025] Open
Abstract
The liver performs crucial roles in energy metabolism, detoxification, and immune regulation. Hepatic diseases, including hepatitis, liver fibrosis, and liver cancer, have posed a significant threat to global health, emphasizing the critical need for the development of novel and effective treatment approaches. Nanotechnology, an emerging technology, has been extensively researched in medicine. Among the many types of nanomaterials, polymeric nanoparticles (NPs) are widely used in drug delivery systems. Compared to traditional therapies, they offer significant advantages in the treatment of liver disease by improving outcomes and reducing side effects. This review introduced the development of liver disease and discussed the application of natural polymers and synthetic polymers in their management. Furthermore, this paper reviewed the application of polymeric nanoparticles -mainly chitosan (CS), hyaluronic acid (HA), polyethylene glycol (PEG) and poly (lactic-co-glycolic acid) (PLGA)-in liver disease treatment, focusing on their use in various delivery systems for pure bioactive compounds of natural origin, drugs, nucleic acids, peptides, and others. Finally, the challenges and future perspectives of the NPs were discussed to provide guidance for further research directions, with the aim of promoting the clinical application of nanotherapeutics in treating hepatic diseases.
Collapse
Affiliation(s)
| | | | - Xinyu Li
- Clinical Laboratory of China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
7
|
Eaton DA, Lynn AY, Surprenant JM, Deschenes EI, Guerra ME, Rivero R, Yung NK, O’Connor M, Glazer PM, Bahtiyar MO, Saltzman WM, Stitelman DH. Biodistribution of Polymeric Nanoparticles following in utero Delivery to a Nonhuman Primate. Biomed Hub 2025; 10:23-32. [PMID: 39845408 PMCID: PMC11753793 DOI: 10.1159/000543138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Monogenic diseases can be diagnosed before birth. Systemic fetal administration of nanoparticles (NPs) grants therapeutic access to developing stem cell populations impacted by these classes of disease. Delivery of editing reagents in these NPs administered before birth has yielded encouraging results in preclinical mouse models of monogenic diseases. Methods To translate this strategy clinically, the safety and efficacy of this strategy in larger animals will be necessary. We performed a pilot biodistribution study in 3 fetal nonhuman primates (NHPs) in mid-gestation examining systemic delivery of polymeric NPs loaded with fluorescent dye. Results We found several similarities in distribution to our experience in mice, namely, extensive uptake in fetal liver and spleen. A striking finding that is not recapitulated in the mouse was the accumulation of NPs in the zones of proliferation and ossification of the fetal bone. Of great importance, there did not appear to be NP accumulation in the fetal male or female germline zones or maternal tissue. Conclusion These studies were vital to the next step of testing editing reagents in the fetal NHP with a goal of treating monogenic diseases before birth.
Collapse
Affiliation(s)
- David A. Eaton
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
- Departments of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Anna Y. Lynn
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Emily I. Deschenes
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
- Departments of Biomedical Engineering, Yale University, New Haven, CT, USA
| | | | - Rachel Rivero
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Nicholas K. Yung
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Peter M. Glazer
- Departments of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
- Departments of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Mert Ozan Bahtiyar
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - W. Mark Saltzman
- Departments of Biomedical Engineering, Yale University, New Haven, CT, USA
- Departments of Chemical and Environmental Engineering, Yale University, New Haven, CT, USA
- Departments of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
- Departments of Dermatology, Yale University, New Haven, CT, USA
| | - David H. Stitelman
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
8
|
Szachniewicz MM, Neustrup MA, van den Eeden SJF, van Meijgaarden KE, Franken KLMC, van Veen S, Koning RI, Limpens RWAL, Geluk A, Bouwstra JA, Ottenhoff THM. Evaluation of PLGA, lipid-PLGA hybrid nanoparticles, and cationic pH-sensitive liposomes as tuberculosis vaccine delivery systems in a Mycobacterium tuberculosis challenge mouse model - A comparison. Int J Pharm 2024; 666:124842. [PMID: 39424087 DOI: 10.1016/j.ijpharm.2024.124842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Tuberculosis (TB) continues to pose a global threat for millennia, currently affecting over 2 billion people and causing 10.6 million new cases and 1.3 million deaths annually. The only existing vaccine, Mycobacterium Bovis Bacillus Calmette-Guérin (BCG), provides highly variable and inadequate protection in adults and adolescents. This study explores newly developed subunit tuberculosis vaccines that use a multistage protein fusion antigen Ag85b-ESAT6-Rv2034 (AER). The protection efficacy, as well as in vivo induced immune responses, were compared for five vaccines: BCG; AER-CpG/MPLA mix; poly(D,L-lactic-co-glycolic acid) (PLGA); lipid-PLGA hybrid nanoparticles (NPs); and cationic pH-sensitive liposomes (the latter three delivering AER together with CpG and MPLA). All vaccines, except the AER-adjuvant mix, induced protection in Mycobacterium tuberculosis (Mtb)-challenged C57/Bl6 mice as indicated by a significant reduction in bacterial burden in lungs and spleens of the animals. Four AER-based vaccines significantly increased the number of circulating multifunctional CD4+ and CD8+ T-cells producing IL-2, IFNγ, and TNFα, exhibiting a central memory phenotype. Furthermore, AER-based vaccines induced an increase in CD69+ B-cell counts as well as high antigen-specific antibody titers. Unexpectedly, none of the observed immune responses were associated with the bacterial burden outcome, such that the mechanism responsible for the observed vaccine-induced protection of these vaccines remains unclear. These findings suggest the existence of non-classical protective mechanisms for Mtb infection, which could, once identified, provide interesting targets for novel vaccines.
Collapse
Affiliation(s)
- Mikołaj M Szachniewicz
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands.
| | - Malene A Neustrup
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - Susan J F van den Eeden
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Krista E van Meijgaarden
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Kees L M C Franken
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Suzanne van Veen
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Roman I Koning
- Electron Microscopy Facility, Leiden University Medical Center (LUMC), the Netherlands
| | - Ronald W A L Limpens
- Electron Microscopy Facility, Leiden University Medical Center (LUMC), the Netherlands
| | - Annemieke Geluk
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| | - Joke A Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - Tom H M Ottenhoff
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), the Netherlands
| |
Collapse
|
9
|
Keshavarz Shahbaz S, Koushki K, Izadi O, Penson PE, Sukhorukov VN, Kesharwani P, Sahebkar A. Advancements in curcumin-loaded PLGA nanoparticle delivery systems: progressive strategies in cancer therapy. J Drug Target 2024; 32:1207-1232. [PMID: 39106154 DOI: 10.1080/1061186x.2024.2389892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
Cancer is a leading cause of death worldwide, and imposes a substantial socioeconomic burden with little impact especially on aggressive types of cancer. Conventional therapies have many serious side effects including generalised systemic toxicity which limits their long-term use. Tumour resistance and recurrence is another main problem associated with conventional therapy. Purified or extracted natural products have been investigated as cost-effective cancer chemoprotective agents with the potential to reverse or delaying carcinogenesis. Curcumin (CUR) as a natural polyphenolic component, exhibits many pharmacological activities such as anti-cancer, anti-inflammatory, anti-microbial, activity against neurodegenerative diseases including Alzheimer, antidiabetic activities (type II diabetes), anticoagulant properties, wound healing effects in both preclinical and clinical studies. Despite these effective protective properties, CUR has several limitations, including poor aqueous solubility, low bioavailability, chemical instability, rapid metabolism and a short half-life time. To overcome the pharmaceutical problems associated with free CUR, novel nanomedicine strategies (including polymeric nanoparticles (NPs) such as poly (lactic-co-glycolic acid) (PLGA) NPs have been developed. These formulations have the potential to improve the therapeutic efficacy of curcuminoids. In this review, we comprehensively summarise and discuss recent in vitro and in vivo studies to explore the pharmaceutical significance and clinical benefits of PLGA-NPs delivery system to improve the efficacy of CUR in the treatment of cancer.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Khadijeh Koushki
- Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Omid Izadi
- Department of Industrial Engineering, ACECR Institute of Higher Education Kermanshah, Kermanshah, Iran
| | - Peter E Penson
- Clinical Pharmacy and Therapeutics Research Group, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
- Liverpool Centre for Cardiovascular Science, Liverpool, UK
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Zhang S, Fang H, Tian H. Recent Advances in Degradable Biomedical Polymers for Prevention, Diagnosis and Treatment of Diseases. Biomacromolecules 2024; 25:7015-7057. [PMID: 39420482 DOI: 10.1021/acs.biomac.4c01193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Biomedical polymers play a key role in preventing, diagnosing, and treating diseases, showcasing a wide range of applications. Their unique advantages, such as rich source, good biocompatibility, and excellent modifiability, make them ideal biomaterials for drug delivery, biomedical imaging, and tissue engineering. However, conventional biomedical polymers suffer from poor degradation in vivo, increasing the risks of bioaccumulation and potential toxicity. To address these issues, degradable biomedical polymers can serve as an alternative strategy in biomedicine. Degradable biomedical polymers can efficiently relieve bioaccumulation in vivo and effectively reduce patient burden in disease management. This review comprehensively introduces the classification and properties of biomedical polymers and the recent research progress of degradable biomedical polymers in various diseases. Through an in-depth analysis of their classification, properties, and applications, we aim to provide strong guidance for promoting basic research and clinical translation of degradable biomedical polymers.
Collapse
Affiliation(s)
- Siting Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
11
|
Bezir K, Pelit Arayici P, Akgül B, Abamor EŞ, Acar S. RABV antigenic peptide loaded polymeric nanoparticle production, characterization, and preliminary investigation of its biological activity. NANOTECHNOLOGY 2024; 36:025603. [PMID: 39383880 DOI: 10.1088/1361-6528/ad84fe] [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: 07/29/2024] [Accepted: 10/09/2024] [Indexed: 10/11/2024]
Abstract
Nanoparticle-based antigen carrier systems have become a significant area of research with the advancement of nanotechnology. Biodegradable polymers have emerged as particularly promising carrier vehicles due to their ability to address the limitations of existing vaccine systems. In this study, we successfully encapsulated the G5-24 linear peptide, located between amino acids 253 and 275 in the primary sequence of the rabies virus G protein, into biodegradable and biocompatible PLGA copolymer using the double emulsion solvent evaporation method. The resulting nanoparticles had a size of approximately 230.9 ± 0.9074 nm, with a PDI value of 0.168 ± 0.017 and a zeta potential value of -9.86 ± 0.132 mV. SEM images confirmed that the synthesized nanoparticles were uniform in size and distribution. Additionally, FTIR spectra indicated successful peptide loading into the nanoparticles. The encapsulation efficiency of the peptide-loaded nanoparticles was 73.3%, with a peptide loading capacity of 48.2% and a reaction yield of 30.4%. Peptide release studies demonstrated that 65.55% of the peptide was released in a controlled manner over 28 d, following a 'biphasic burst release' profile consistent with the degradation profile of PLGA. This controlled release is particularly beneficial for vaccine studies. Cytotoxicity tests revealed that the R-NP formulation did not induce cytotoxicity in fibroblast cells and enhanced NO production in macrophages, indicating its potential for vaccine development.
Collapse
Affiliation(s)
- Kübra Bezir
- Faculty of Engineering and Natural Sciences, Bioengineering Department, Bursa Technical University, Bursa, Turkey
| | - Pelin Pelit Arayici
- Faculty of Chemical and Metallurgical Engineering, Bioengineering Department, Yildiz Technical University, Istanbul, Turkey
- Health Biotechnology Joint Research and Application Center of Excellence, 34220 Esenler, Istanbul, Turkey
| | - Buşra Akgül
- Faculty of Chemical and Metallurgical Engineering, Bioengineering Department, Yildiz Technical University, Istanbul, Turkey
| | - Emrah Şefik Abamor
- Faculty of Chemical and Metallurgical Engineering, Bioengineering Department, Yildiz Technical University, Istanbul, Turkey
| | - Serap Acar
- Faculty of Chemical and Metallurgical Engineering, Bioengineering Department, Yildiz Technical University, Istanbul, Turkey
| |
Collapse
|
12
|
Song Q, Li J, Li T, Li H. Nanomaterials that Aid in the Diagnosis and Treatment of Alzheimer's Disease, Resolving Blood-Brain Barrier Crossing Ability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403473. [PMID: 39101248 PMCID: PMC11481234 DOI: 10.1002/advs.202403473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/04/2024] [Indexed: 08/06/2024]
Abstract
As a form of dementia, Alzheimer's disease (AD) suffers from no efficacious cure, yet AD treatment is still imperative, as it ameliorates the symptoms or prevents it from deteriorating or maintains the current status to the longest extent. The human brain is the most sensitive and complex organ in the body, which is protected by the blood-brain barrier (BBB). This yet induces the difficulty in curing AD as the drugs or nanomaterials that are much inhibited from reaching the lesion site. Thus, BBB crossing capability of drug delivery system remains a significant challenge in the development of neurological therapeutics. Fortunately, nano-enabled delivery systems possess promising potential to achieve multifunctional diagnostics/therapeutics against various targets of AD owing to their intriguing advantages of nanocarriers, including easy multifunctionalization on surfaces, high surface-to-volume ratio with large payloads, and potential ability to cross the BBB, making them capable of conquering the limitations of conventional drug candidates. This review, which focuses on the BBB crossing ability of the multifunctional nanomaterials in AD diagnosis and treatment, will provide an insightful vision that is conducive to the development of AD-related nanomaterials.
Collapse
Affiliation(s)
- Qingting Song
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Junyou Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Ting Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Hung‐Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
13
|
Gonzalez-Melero L, Santos-Vizcaino E, Varela-Calvino R, Gomez-Tourino I, Asumendi A, Boyano MD, Igartua M, Hernandez RM. PLGA-PEI nanoparticle covered with poly(I:C) for personalised cancer immunotherapy. Drug Deliv Transl Res 2024; 14:2788-2803. [PMID: 38427275 PMCID: PMC11525302 DOI: 10.1007/s13346-024-01557-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Melanoma is the main cause of death among skin cancers and its incidence worldwide has been experiencing an appalling increase. However, traditional treatments lack effectiveness in advanced or metastatic patients. Immunotherapy, meanwhile, has been shown to be an effective treatment option, but the rate of cancers responding remains far from ideal. Here we have developed a personalized neoantigen peptide-based cancer vaccine by encapsulating patient derived melanoma neoantigens in polyethylenimine (PEI)-functionalised poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) and coating them with polyinosinic:polycytidylic acid (poly(I:C)). We found that PLGA NPs can be effectively modified to be coated with the immunoadjuvant poly(I:C), as well as to encapsulate neoantigens. In addition, we found that both dendritic cells (DCs) and lymphocytes were effectively stimulated. Moreover, the developed NP was found to have a better immune activation profile than NP without poly(I:C) or without antigen. Our results demonstrate that the developed vaccine has a high capacity to activate the immune system, efficiently maturing DCs to present the antigen of choice and promoting the activity of lymphocytes to exert their cytotoxic function. Therefore, the immune response generated is optimal and specific for the elimination of melanoma tumour cells.
Collapse
Affiliation(s)
- Lorena Gonzalez-Melero
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
| | - Ruben Varela-Calvino
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Santiago de Compostela, Santiago, Spain
| | - Iria Gomez-Tourino
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Santiago, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
| | - Aintzane Asumendi
- Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Maria Dolores Boyano
- Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
14
|
Qi W, Yu Y, Yang C, Wang X, Jiang Y, Zhang L, Yu Z. Nanospheres as the delivery vehicle: novel application of Toxoplasma gondii ribosomal protein S2 in PLGA and chitosan nanospheres against acute toxoplasmosis. Front Immunol 2024; 15:1475280. [PMID: 39416787 PMCID: PMC11480959 DOI: 10.3389/fimmu.2024.1475280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Toxoplasma gondii (T. gondii) is a zoonotic disease that poses great harm to humans and animals. So far, no effective T. gondii vaccine has been developed to provide fully protection against such parasites. Recently, numerous researches have focused on the use of poly-lactic-co-glycolic acid (PLGA) and chitosan (CS) for the vaccines against T. gondii infections. In this study, we employed PLGA and CS as the vehicles for T. gondii ribosome protein (TgRPS2) delivery. TgRPS2-PLGA and TgRPS2-CS nanospheres were synthesized by double emulsion solvent evaporation and ionic gelation technique as the nano vaccines. Before immunization in animals, the release efficacy and toxicity of the synthesized nanospheres were evaluated in vitro. Then, ICR mice were immunized intramuscularly, and immune protections of the synthesized nanospheres were assessed. The results showed that TgRPS2-PLGA and TgRPS2-CS nanospheres could induce higher levels of IgG and cytokines, activate dendritic cells, and promote the expression of histocompatibility complexes. The splenic lymphocyte proliferation and the enhancement in the proportion of CD4+ and CD8+ T lymphocytes were also observed in immunized animals. In addition, two types of nanospheres could significantly inhabit the replications of T. gondii in cardiac muscles and spleen tissues. All these obtained results in this study demonstrated that the TgRPS2 protein delivered by PLGA or CS nanospheres provided satisfactory immunoprotective effects in resisting T. gondii, and such formulations illustrated potential as prospective preventive agents for toxoplasmosis.
Collapse
Affiliation(s)
- WeiYu Qi
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - YouLi Yu
- Institute of Animal Science, Ningxia Academy of Agricultural and Forestry Science, Yinchuan, China
| | - ChenChen Yang
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - XiaoJuan Wang
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - YuChen Jiang
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - Li Zhang
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| | - ZhengQing Yu
- College of animal science and technology, Ningxia University, Yinchuan, Ningxia, China
| |
Collapse
|
15
|
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] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [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)
| | | | | | - 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
|
16
|
Zhou Y, Wei R, Wang L, Li J, Wang W, Jiang G, Tan S, Li F, Wang X, Ma X, Xi L. Tumor targeting peptide TMTP1 modified Antigen capture Nano-vaccine combined with chemotherapy and PD-L1 blockade effectively inhibits growth of ovarian cancer. J Nanobiotechnology 2024; 22:483. [PMID: 39138475 PMCID: PMC11320875 DOI: 10.1186/s12951-024-02744-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
The mortality of ovarian cancer (OC) has long been the highest among gynecological malignancies. Although OC is considered to be an immunogenic tumor, the effect of immunotherapy is not satisfactory. The immunosuppressive microenvironment is one reason for this, and the absence of recognized effective antigens for vaccines is another. Chemotherapy, as one of the most commonly used treatment for OC, can produce chemotherapy-associated antigens (CAAs) during treatment and show the effect of in situ vaccine. Herein, we designed an antigen capture nano-vaccine NP-TP1@M-M with tumor targeting peptide TMTP1 and dendritic cell (DC) receptor mannose assembled on the surface and adjuvant monophosphoryl lipid A (MPLA) encapsulated in the core of poly (D, L-lactide-co-glycolide) (PLGA) nanoparticles. PLGA itself possessed the ability of antigen capture. TMTP1 was a tumor-homing peptide screened by our research team, which held extensive and excellent tumor targeting ability. After these modifications, NP-TP1@M-M could capture and enrich more tumor-specific antigens after chemotherapy, stimulate DC maturation, activate the adaptive immunity and combined with immune checkpoint blockade to maximize the release of the body's immune potential, providing an eutherapeutic strategy for the treatment of OC.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Wei
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Wang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jie Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guiying Jiang
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, China
| | - Songwei Tan
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xueqian Wang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyi Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ling Xi
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
17
|
La Manna S, Cugudda A, Mercurio FA, Leone M, Fortuna S, Di Natale C, Lagreca E, Netti PA, Panzetta V, Marasco D. PEGylated SOCS3 Mimetics Encapsulated into PLGA-NPs as Selective Inhibitors of JAK/STAT Pathway in TNBC Cells. Int J Nanomedicine 2024; 19:7237-7251. [PMID: 39050870 PMCID: PMC11268778 DOI: 10.2147/ijn.s441205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 05/28/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction SOCS3 (suppressor of cytokine signaling 3) protein is a crucial regulator of cytokine-induced inflammation, and its administration has been shown to have therapeutic effects. Recently, we designed a chimeric proteomimetic of SOCS3, mimicking the interfacing regions of a ternary complex composed of SOCS3, JAK2 (Janus kinase 2) and gp130 (glycoprotein 130) proteins. The derived chimeric peptide, KIRCONG chim, demonstrated limited mimetic function owing to its poor water solubility. Methods We report investigations concerning a PEGylated variant of KIRCONG mimetic, named KIRCONG chim, bearing a PEG (Polyethylene glycol) moiety as a linker of noncontiguous SOCS3 regions. Its ability to bind to the catalytic domain of JAK2 was evaluated through MST (MicroScale Thermophoresis), as well as its stability in biological serum assays. The structural features of the cyclic compounds were investigated by CD (circular dichroism), nuclear magnetic resonance (NMR), and molecular dynamic (MD) studies. To evaluate the cellular effects, we employed a PLGA-nanoparticle as a delivery system after characterization using DLS and SEM techniques. Results KIRCONG chim PEG-revealed selective penetration into triple-negative breast cancer (TNBC) MDA-MB-231 cells with respect to the human breast epithelial cell line (MCF10A), acting as a potent inhibitor of STAT3 phosphorylation. Discussion Overall, the data indicated that miniaturization of the SOCS3 protein is a promising therapeutic approach for aberrant dysregulation of JAK/STAT during cancer progression.
Collapse
Affiliation(s)
- Sara La Manna
- Department of Pharmacy, CIRPEB: Research Center on Bioactive Peptides- University of Naples Federico II, Naples, 80131, Italy
| | - Alessia Cugudda
- Department of Pharmacy, CIRPEB: Research Center on Bioactive Peptides- University of Naples Federico II, Naples, 80131, Italy
| | | | - Marilisa Leone
- Institute of Biostructures and Bioimaging (CNR), Naples, 80131, Italy
| | - Sara Fortuna
- Italian Institute of Technology (IIT), Genova, 16152, Italy
| | - Concetta Di Natale
- Department of Ingegneria Chimica del Materiali e della Produzione Industriale (DICMAPI), University of Naples Federico II, Naples, 80125, Italy
| | - Elena Lagreca
- Department of Ingegneria Chimica del Materiali e della Produzione Industriale (DICMAPI), University of Naples Federico II, Naples, 80125, Italy
| | - Paolo Antonio Netti
- Department of Ingegneria Chimica del Materiali e della Produzione Industriale (DICMAPI), University of Naples Federico II, Naples, 80125, Italy
| | - Valeria Panzetta
- Department of Ingegneria Chimica del Materiali e della Produzione Industriale (DICMAPI), University of Naples Federico II, Naples, 80125, Italy
| | - Daniela Marasco
- Department of Pharmacy, CIRPEB: Research Center on Bioactive Peptides- University of Naples Federico II, Naples, 80131, Italy
| |
Collapse
|
18
|
Liu K, Peng J, Guo Y, Li Y, Qi X, Duan D, Li T, Li J, Niu Y, Han G, Zhao Y. Expanding the Potential of Neoantigen Vaccines: Harnessing Bacille Calmette-Guérin Cell-Wall-Based Nanoscale Adjuvants for Enhanced Cancer Immunotherapy. ACS NANO 2024; 18:11910-11920. [PMID: 38680054 DOI: 10.1021/acsnano.4c01691] [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: 05/01/2024]
Abstract
Personalized antitumor immunotherapy utilizing neoantigen vaccines holds great promise. However, the limited immunogenicity of existing recognized neoantigens and the inadequate stimulation of antitumor immune responses by conventional adjuvants pose significant challenges. To address these limitations, we developed a nanovaccine that combines a BCG bacterial cell wall skeleton (BCG-CWS) based nanoscale adjuvant (BCNA) with peptide neoantigens (M27 and M30). This integrated approach provides an efficient translational strategy for cancer immunotherapy. The BCNA nanovaccine, formulated with PLGA as an emulsifier, exhibits excellent biocompatibility and superior antigen presentation compared with conventional BCG-CWS adjuvants. Subcutaneous immunization with the BCNA-based nanovaccine effectively targets lymph nodes, eliciting robust innate and tumor-specific immune responses. Importantly, our findings demonstrate that BCNAs significantly enhance neoantigen immunogenicity while minimizing acute systemic toxicity. Furthermore, when combined with a mouse PD-L1 antibody, our strategy achieves complete tumor elimination in 60% of cases and prevents 25% of tumor growth in a melanoma mouse model. In conclusion, our BCNA-based nanovaccine represents a promising avenue for advancing personalized therapeutic neoantigen vaccines and holds significant implications for enhancing personalized immunotherapy and improving patient outcomes in the field of cancer treatment.
Collapse
Affiliation(s)
- Kangkang Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jing Peng
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
- Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, 364 Plantation Street, LRB 806, Worcester, Massachusetts 01605, United States
| | - Yunfei Guo
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yiming Li
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xiang Qi
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Dengyi Duan
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Taipeng Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jianmin Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yuanjie Niu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Gang Han
- Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, 364 Plantation Street, LRB 806, Worcester, Massachusetts 01605, United States
| | - Yang Zhao
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
- Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, 364 Plantation Street, LRB 806, Worcester, Massachusetts 01605, United States
| |
Collapse
|
19
|
Xiong Y, Liu Z, Wang Y, Wang J, Zhou X, Li X. Development and Evaluation of a Water-Free In Situ Depot Gel Formulation for Long-Acting and Stable Delivery of Peptide Drug ACTY116. Pharmaceutics 2024; 16:620. [PMID: 38794282 PMCID: PMC11125081 DOI: 10.3390/pharmaceutics16050620] [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: 03/20/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
In situ depot gel is a type of polymeric long-acting injectable (pLAI) drug delivery system; compared to microsphere technology, its preparation process is simpler and more conducive to industrialization. To ensure the chemical stability of peptide ACTY116, we avoided the use of harsh conditions such as high temperatures, high shear mixing, or homogenization; maintaining a water-free and oxygen-free environment was also critical to prevent hydrolysis and oxidation. Molecular dynamics (MDs) simulations were employed to assess the stability mechanism between ACTY116 and the pLAI system. The initial structure of ACTY116 with an alpha helix conformation was constructed using SYBYL-X, and the copolymer PLGA was generated by AMBER 16; results showed that PLGA-based in situ depot gel improved conformational stability of ACTY116 through hydrogen bonds formed between peptide ACTY116 and the components of the pLAI formulation, while PLGA (Poly(DL-lactide-co-glycolide)) also created steric hindrance and shielding effects to prevent conformational changes. As a result, the chemical and conformational stability and in vivo long-acting characteristics of ACTY116 ensure its enhanced efficacy. In summary, we successfully achieved our objective of developing a highly stable peptide-loaded long-acting injectable (LAI) in situ depot gel formulation that is stable for at least 3 months under harsh conditions (40 °C, above body temperature), elucidating the underlying stabilisation mechanism, and the high stability of the ACTY116 pLAI formulation creates favourable conditions for its in vivo pharmacological activity lasting for weeks or even months.
Collapse
Affiliation(s)
- Yingxin Xiong
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing 400038, China;
| | - Zhirui Liu
- Department of Pharmacy, Xinan Hospital, Army Medical University, Chongqing 400038, China;
| | - Yuanqiang Wang
- Chongqing School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (Y.W.); (J.W.)
| | - Jiawei Wang
- Chongqing School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (Y.W.); (J.W.)
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, China
| | - Xiaohui Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing 400038, China;
- Engineering Research Center for Pharmacodynamics Evaluation, College of Pharmacy, Army Medical University, Chongqing 400038, China
| |
Collapse
|
20
|
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
|
21
|
Cassaidy B, Moser BA, Solanki A, Chen Q, Shen J, Gotsis K, Lockhart Z, Rutledge N, Rosenberger MG, Dong Y, Davis D, Esser- Kahn AP. Immune Potentiation of PLGA Controlled-Release Vaccines for Improved Immunological Outcomes. ACS OMEGA 2024; 9:11608-11614. [PMID: 38496947 PMCID: PMC10938429 DOI: 10.1021/acsomega.3c06552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/25/2024] [Accepted: 02/08/2024] [Indexed: 03/19/2024]
Abstract
With the emergence of SARS-CoV-2 and the continued emergence of new infectious diseases, there is a need to improve and expand current vaccine technology. Controlled-release subunit vaccines provide several benefits over current vaccines on the market, including the use of less antigen and fewer boost doses. Previously, our group reported molecules that alter NF-κB signaling improved the vaccine's performance and improved adjuvant-related tolerability. In this report, we test how these immune potentiators will influence responses when included as part of a controlled-release poly(lactic-co-glycolic) vaccine formulation. Murine in vivo studies revealed that SN50 and honokiol improved antibody levels at early vaccine time points. Microparticles with SN50 produced strong antibody levels over a longer period compared to microparticles without SN50. The same particles also increased T-cell activity. All of the immune potentiators tested further promoted Th2 humoral responses already exhibited by the control CpG OVA microparticle formulation. Overall, under controlled-release conditions, immune potentiators enhance the existing effects of controlled-release formulations, making it a potentially beneficial additive for controlled-release vaccine formulations.
Collapse
Affiliation(s)
- Britteny
J. Cassaidy
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Brittany A. Moser
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Ani Solanki
- Animal
Resource Center, University of Chicago, Chicago, Illinois 60637, United States
| | - Qing Chen
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Jingjing Shen
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Kristen Gotsis
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Zoe Lockhart
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Nakisha Rutledge
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Matthew G. Rosenberger
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Yixiao Dong
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Delaney Davis
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| | - Aaron P. Esser- Kahn
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S. Ellis Avenue, Chicago, Illinois 60637, United States
| |
Collapse
|
22
|
Sunazuka Y, Ueda K, Higashi K, Wada K, Moribe K. Mechanistic Analysis of Temperature-Dependent Curcumin Release from Poly(lactic-co-glycolic acid)/Poly(lactic acid) Polymer Nanoparticles. Mol Pharm 2024; 21:1424-1435. [PMID: 38324797 DOI: 10.1021/acs.molpharmaceut.3c01066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
In this study, we investigated the mechanism of curcumin (CUR) release from poly(lactic-co-glycolic acid) (PLGA) and poly(lactic acid) (PLA) nanoparticles (NPs) by evaluating the temperature-dependent CUR release. NPs were prepared by the nanoprecipitation method using various PLGA/PLA polymers with different lactic:glycolic ratios (L:G ratios) and molecular weights. Increasing the polymer molecular weight resulted in a decrease in the particle size of NPs. The wet glass transition temperature (Tg) of PLGA/PLA NPs was lower than the intrinsic polymer Tg, which can be derived from the water absorption and nanosizing of the polymer. The reduction in Tg was more significant for the PLGA/PLA NPs with lower polymer L:G ratios and lower polymer molecular weight. The greater decrease of Tg in the lower polymer L:G ratios was possibly caused by the higher water absorption due to the more hydrophilic nature of the glycolic acid segment than that of the lactic acid segment. The efficient water absorption in PLGA/PLA NPs with lower molecular weight could cause a significant reduction of Tg as it has lower hydrophobicity. CUR release tests from the PLGA/PLA NPs exhibited enhanced CUR release with increasing temperatures, irrespective of polymer species. By fitting the CUR release profiles into mathematical models, the CUR release process was well described by an initial burst release followed by a diffusion-controlled release. The wet Tg and particle size of the PLGA/PLA NPs affected the amount and temperature dependence of the initial burst release of CUR. Above the wet Tg of NPs, the initial burst release of CUR increased sharply. Smaller particle sizes of PLGA/PLA NPs led to a higher fraction of initial CUR burst release, which was more pronounced above the wet Tg of NPs. The wet Tg and particle sizes of the PLGA/PLA NPs also influenced the diffusion-controlled CUR release. The diffusion rate of CUR in the NPs increased as the wet Tg values of the NPs decreased. The diffusion path length of CUR was affected by the particle size, with larger particle size resulting in a prolonged diffusion-controlled release of CUR. This study highlighted that for the formulation development of PLGA/PLA NPs, suitable PLGA/PLA polymers should be selected considering the physicochemical properties of PLGA/PLA NPs and their correlation with the release behavior of encapsulated drugs at the application temperature.
Collapse
Affiliation(s)
- Yushi Sunazuka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
- Nippon Boehringer Ingelheim Co. Ltd., 6-7-5 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kenjirou Higashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Koichi Wada
- Nippon Boehringer Ingelheim Co. Ltd., 6-7-5 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kunikazu Moribe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
23
|
Wang X, Wu H, Fang C, Li Z. Insights into innate immune cell evasion by Chlamydia trachomatis. Front Immunol 2024; 15:1289644. [PMID: 38333214 PMCID: PMC10850350 DOI: 10.3389/fimmu.2024.1289644] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Chlamydia trachomatis, is a kind of obligate intracellular pathogen. The removal of C. trachomatis relies primarily on specific cellular immunity. It is currently considered that CD4+ Th1 cytokine responses are the major protective immunity against C. trachomatis infection and reinfection rather than CD8+ T cells. The non-specific immunity (innate immunity) also plays an important role in the infection process. To survive inside the cells, the first process that C. trachomatis faces is the innate immune response. As the "sentry" of the body, mast cells attempt to engulf and remove C. trachomatis. Dendritic cells present antigen of C. trachomatis to the "commanders" (T cells) through MHC-I and MHC-II. IFN-γ produced by activated T cells and natural killer cells (NK) further activates macrophages. They form the body's "combat troops" and produce immunity against C. trachomatis in the tissues and blood. In addition, the role of eosinophils, basophils, innate lymphoid cells (ILCs), natural killer T (NKT) cells, γδT cells and B-1 cells should not be underestimated in the infection of C. trachomatis. The protective role of innate immunity is insufficient, and sexually transmitted diseases (STDs) caused by C. trachomatis infections tend to be insidious and recalcitrant. As a consequence, C. trachomatis has developed a unique evasion mechanism that triggers inflammatory immunopathology and acts as a bridge to protective to pathological adaptive immunity. This review focuses on the recent advances in how C. trachomatis evades various innate immune cells, which contributes to vaccine development and our understanding of the pathophysiologic consequences of C. trachomatis infection.
Collapse
Affiliation(s)
| | | | | | - Zhongyu Li
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, School of Nursing, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
24
|
Sheikhi M, Sharifzadeh M, Hennink WE, Firoozpour L, Hajimahmoodi M, Khoshayand MR, Shirangi M. Design of experiments approach for the development of a validated method to determine the exenatide content in poly(lactide-co-glycolide) microspheres. Eur J Pharm Biopharm 2023; 192:56-61. [PMID: 37783361 DOI: 10.1016/j.ejpb.2023.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/20/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
Due to the lack of pharmacopeia guidelines for injectable microspheres based on poly (D, L-lactide-co-glycolide) (PLGA), an internal method validation is a critical prerequisite for quality assurance. One of the essential issues of developing peptide-based drugs loaded PLGA microspheres is the precise determination of the amount of peptide drug entrapped in the microspheres. The aim of this study is the development and optimization of a method for measuring the drug content loading of PLGA microspheres using exenatide as a model peptide drug. Exenatide-loaded PLGA microspheres were prepared by a double emulsion solvent evaporation method. The extraction method to determine exenatide content in microspheres was optimized using Design of Experiments (DoE) approach. After the initial screening of six factors, using Fractional Factorial design (FFD), four of them, including type of organic solvent, buffer/organic solvent ratio (v/v), shaking time and pH, exhibited significant effects on the response, namely the exenatide loading, and a Box-Behnken design (BBD) was subsequently applied to obtain its optimum level. The optimum level for organic solvent volume, buffer/organic solvent ratio, shaking time, and pH were 4 ml, 1, 5.6 hrs, and pH 6, respectively. The exenatide content in microspheres under these conditions was 6.4 ± 0.0 (%w/w), whereas a value of 6.1% was predicted by the derived equation. This excellent agreement between the actual and the predicted value demonstrates that the fitted model can thus be used to determine the exenatide content.
Collapse
Affiliation(s)
- Mojgan Sheikhi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Science Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Mannan Hajimahmoodi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Science Tehran, Iran
| | - Mohammad Reza Khoshayand
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Science Tehran, Iran.
| | - Mehrnoosh Shirangi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Science Tehran, Iran.
| |
Collapse
|
25
|
Sim Y, Seong J, Lee S, Kim D, Choi E, Lah MS, Ryu JH. Metal-Organic Framework-Supported Catalase Delivery for Enhanced Photodynamic Therapy via Hypoxia Mitigation. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37883653 DOI: 10.1021/acsami.3c13395] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Tumor hypoxia poses a significant challenge in photodynamic therapy (PDT), which uses molecular oxygen to produce reactive oxygen species upon light excitation of a photosensitizer. For hypoxia mitigation, an enzyme catalase (CAT) can be beneficially used to convert intracellular hydrogen peroxide to molecular oxygen, but its utility is significantly limited due to the intrinsic membrane impermeability. Herein, we present direct integration of CAT into the outer surface of unmodified metal-organic framework (MOF) nanoparticles (NPs) via supramolecular interactions for effective cellular entry of CAT and consequent enhancement of PDT. The results demonstrated that CAT-loaded MOF NPs could successfully enter hypoxic cancer cells, after which the intracellularly delivered CAT molecules became dissociated from the MOF surface to efficiently initiate the oxygen generation and PDT process along with a co-delivered photosensitizer IR780. This achievement suggests that our protein-MOF integration strategy holds great potential in biomedical studies to overcome tumor hypoxia as well as to efficiently deliver biomolecular cargos.
Collapse
Affiliation(s)
- Youjung Sim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Junmo Seong
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seonghwan Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Dohyun Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eunshil Choi
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Myoung Soo Lah
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
26
|
Chen J, Wang H, Zhang L, Yan W, Sheng R. Facile preparation of PEGylated polyethylenimine polymers as vaccine carriers with reduced cytotoxicity and enhanced Interleukin-2 (IL-2) production. Colloids Surf B Biointerfaces 2023; 230:113520. [PMID: 37619373 DOI: 10.1016/j.colsurfb.2023.113520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/26/2023]
Abstract
Developing low-cost, easy-to-prepare, biocompatible and highly efficient vaccine carriers is a promising approach to realize practical cancer immunotherapy. In this study, through facile modification of mPEG5k-4-toluenesulfonate (mPEG5k-OTs) on PEI25k under mild conditions, a series of "stealth" mPEG5k-PEI25k polymers (PP1, PP2 and PP3) were prepared, their structures and physicochemical properties were characterized and theoretically analyzed. The polymers could bind/load ovalbumin (OVA) to form mPEG5k-PEI25k/OVA complexes as negatively charged nanoparticles with small hydrodynamic particle size (80-210 nm) and narrow size distribution. Compared to PEI25k/OVA, lower cytotoxicity could be achieved on mPEG5k-PEI25k/OVA complexes in dendritic cells (DCs). In DCs-RF 33.70 T-cells co-culture system, the mPEG5k-PEI25k/OVA complexes could bring about higher IL-2 production /secretion than that of PEI25k/OVA, notably, the optimum IL-2 secretion could reach 9.3-folds of the PEI25k/OVA under serum condition (10% FBS). Moreover, the cell biological features could be optimized by selecting suitable mPEG5k-grafting ratios and/or mPEG5k-PEI25k/OVA weight ratios. Intracellular imaging results showed that the mPEG5k-PEI25k(PP3)/Rhodamine-OVA complexes mainly localized inside lysosomes. Taken together, this work provided a facile method to prepare "stealth" PEGylated-PEI25k polymers with reduced cytotoxicity, promoted OVA cross-presentation efficiency and improved serum compatibility towards cancer immunotherapy.
Collapse
Affiliation(s)
- Jian Chen
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road 800, Shanghai 200240, China.
| | - Hui Wang
- School of Pharmacy, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Li Zhang
- Instrumental Analysis Center, Shanghai Jiao Tong University, Dongchuan Road, Shanghai 200240, China.
| | - Wanying Yan
- School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road 800, Shanghai 200240, China
| | - Ruilong Sheng
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| |
Collapse
|
27
|
Liu Q, Hu Y, Zheng P, Yang Y, Fu Y, Yang Y, Duan B, Wang M, Li D, Li W, He J, Zheng X, Long Q, Ma Y. Exploiting immunostimulatory mechanisms of immunogenic cell death to develop membrane-encapsulated nanoparticles as a potent tumor vaccine. J Nanobiotechnology 2023; 21:326. [PMID: 37684628 PMCID: PMC10492316 DOI: 10.1186/s12951-023-02031-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/28/2023] [Indexed: 09/10/2023] Open
Abstract
Vaccine is one of the most promising strategies for cancer immunotherapy; however, there are no therapeutic cancer vaccine achieving significant clinical efficacy till now. The main limiting factors include the immune suppression and escape mechanisms developed by tumor and not enough capacity of vaccines to induce a vigorous anti-tumor immunity. This study aimed to develop a strategy of membrane-based biomimetic nanovaccine and investigate the immunological outcomes of utilizing the unique immunostimulatory mechanisms derived of immunogenic cell death (ICD) and of fulfilling a simultaneous nanoscale delivery of a highlighted tumor antigen and broad membrane-associated tumor antigens in the vaccine design. TC-1 tumor cells were treated in vitro with a mixture of mitoxantrone and curcumin for ICD induction, and then chitosan (CS)-coated polylactic co-glycolic acid (PLGA) nanoparticles loaded with HPV16 E744-62 peptides were decorated with the prepared ICD tumor cell membrane (IM); further, the IM-decorated nanoparticles along with adenosine triphosphate (ATP) were embedded with sodium alginate (ALG) hydrogel, And then, the immunological features and therapeutic potency were evaluated in vitro and in vivo. The nanovaccine significantly stimulated the migration, antigen uptake, and maturation of DCs in vitro, improved antigen lysosome escape, and promoted the retention at injection site and accumulation in LNs of the tumor antigen in vivo. In a subcutaneously grafted TC-1 tumor model, the therapeutic immunization of nanovaccine elicited a dramatical antitumor immunity. This study provides a strategy for the development of tumor vaccines.
Collapse
Affiliation(s)
- Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- Institute of Medical Biology, Kunming Medical University, Kunming, 650500, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- Institute of Medical Biology, Kunming Medical University, Kunming, 650500, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- Institute of Medical Biology, Kunming Medical University, Kunming, 650500, China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
- School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Qiong Long
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.
- Institute of Medical Biology, Kunming Medical University, Kunming, 650500, China.
- School of Life Sciences, Yunnan University, Kunming, 650091, China.
| |
Collapse
|
28
|
Shakya AK, Nandakumar KS. Polymer Chemistry Defines Adjuvant Properties and Determines the Immune Response against the Antigen or Vaccine. Vaccines (Basel) 2023; 11:1395. [PMID: 37766073 PMCID: PMC10537360 DOI: 10.3390/vaccines11091395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Activation of the immune system is a needed for designing new antigen/drug delivery systems to develop new therapeutics and for developing animal disease models to study the disease pathogenesis. A weak antigen alone is insufficient to activate the immune system. Sometimes, assistance in the form of polymers is needed to control the release of antigens under in vivo conditions or in the form of an adjuvant to activate the immune system efficiently. Many kinds of polymers from different functional groups are suitable as microbial antigens for inducing therapeutic immune responses against infectious diseases at the preclinical level. The choice of the functionality of polymer varies as per the application type. Polymers from the acid and ester groups are the most common types investigated for protein-based antigens. However, electrostatic interaction-displaying polymers like cationic polymers are the most common type for nucleic acid-based antigens. Metal coordination chemistry is commonly used in polymers designed for cancer immunotherapeutic applications to suppress inflammation and induce a protective immune response. Amide chemistry is widely deployed in polymers used to develop antigen-specific disease models like the experimental autoimmune arthritis murine model.
Collapse
Affiliation(s)
| | - Kutty Selva Nandakumar
- Department of Environmental and Biosciences, School of Business, Innovation and Sustainability, Halmstad University, 30118 Halmstad, Sweden
| |
Collapse
|
29
|
Patil S, Vijayanand S, Menon I, Gomes KB, Kale A, Bagwe P, Yacoub S, Uddin MN, D’Souza MJ. Adjuvanted-SARS-CoV-2 Spike Protein-Based Microparticulate Vaccine Delivered by Dissolving Microneedles Induces Humoral, Mucosal, and Cellular Immune Responses in Mice. Pharmaceuticals (Basel) 2023; 16:1131. [PMID: 37631046 PMCID: PMC10457992 DOI: 10.3390/ph16081131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19 continues to cause an increase in the number of cases and deaths worldwide. Due to the ever-mutating nature of the virus, frequent vaccination against COVID-19 is anticipated. Most of the approved SARS-CoV-2 vaccines are administered using the conventional intramuscular route, causing vaccine hesitancy. Thus, there is a need for an effective, non-invasive vaccination strategy against COVID-19. This study evaluated the synergistic effects of a subunit microparticulate vaccine delivered using microneedles. The microparticles encapsulated a highly immunogenic subunit protein of the SARS-CoV-2 virus, such as the spike protein's receptor binding domain (RBD). Adjuvants were also incorporated to enhance the spike RBD-specific immune response. Our vaccination study reveals that a microneedle-based vaccine delivering these microparticles induced spike RBD-specific IgM, IgG, IgG1, IgG2a, and IgA antibodies. The vaccine also generated high levels of CD4+ and CD8a+ molecules in the secondary lymphoid organs. Overall, dissolving microneedles delivery spike RBD antigen in microparticulate form induced a robust immune response, paving the way for an alternative self-administrable, non-invasive vaccination strategy against COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mohammad N. Uddin
- Center for Drug Delivery and Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (S.P.); (S.V.); (I.M.); (K.B.G.); (A.K.); (P.B.); (S.Y.)
| | - Martin J. D’Souza
- Center for Drug Delivery and Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; (S.P.); (S.V.); (I.M.); (K.B.G.); (A.K.); (P.B.); (S.Y.)
| |
Collapse
|
30
|
Chen H, Zheng X, Li L, Huang L, Huang W, Ma Y. Peptide-Based Therapeutic HPV Cancer Vaccine Synthesized via Bacterial Outer Membrane Vesicles. Int J Nanomedicine 2023; 18:4541-4554. [PMID: 37576463 PMCID: PMC10422965 DOI: 10.2147/ijn.s416706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Background Peptide-based vaccines have broad application prospects because of their safety, simple preparation, and effectiveness, especially in the development of personalized cancer vaccines, which have shown great advantages. However, the current peptide-based vaccines often require artificial synthesis and intricate delivery technology, which increases the cost and complexity of preparation. Methods Here, we developed a simple technique for combining a peptide and a delivery system using the natural secretion system of bacteria. Specifically, we biosynthesized an antigenic peptide in bacteria, which was then extracellularly released through the bacterial secretory vesicles, thus simultaneously achieving the biosynthesis and delivery of the peptide. Results The system utilizes the natural properties of bacterial vesicles to promote antigen uptake and dendritic cell (DC) maturation. Therefore, tumor-specific CD4+ Th1 and CD8+ cytotoxic T lymphocyte (CTL) responses were induced in TC-1 tumor-bearing mice, thereby efficiently suppressing tumor growth. Conclusion This research promotes innovation and extends the application of peptide-based vaccine biosynthesis technology. Importantly, it provides a new method for personalized cancer immunotherapy that uses screened peptides as antigens in the future.
Collapse
Affiliation(s)
- Haoqian Chen
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, People’s Republic of China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, People’s Republic of China
- School of Life Sciences, Yunnan University, Kunming, People’s Republic of China
| | - Lingjue Li
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, People’s Republic of China
| | - Lishuxin Huang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming, People’s Republic of China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, People’s Republic of China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, People’s Republic of China
| |
Collapse
|
31
|
Shi M, McHugh KJ. Strategies for overcoming protein and peptide instability in biodegradable drug delivery systems. Adv Drug Deliv Rev 2023; 199:114904. [PMID: 37263542 PMCID: PMC10526705 DOI: 10.1016/j.addr.2023.114904] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
The global pharmaceutical market has recently shifted its focus from small molecule drugs to peptide, protein, and nucleic acid drugs, which now comprise a majority of the top-selling pharmaceutical products on the market. Although these biologics often offer improved drug specificity, new mechanisms of action, and/or enhanced efficacy, they also present new challenges, including an increased potential for degradation and a need for frequent administration via more invasive administration routes, which can limit patient access, patient adherence, and ultimately the clinical impact of these drugs. Controlled-release systems have the potential to mitigate these challenges by offering superior control over in vivo drug levels, localizing these drugs to tissues of interest (e.g., tumors), and reducing administration frequency. Unfortunately, adapting controlled-release devices to release biologics has proven difficult due to the poor stability of biologics. In this review, we summarize the current state of controlled-release peptides and proteins, discuss existing techniques used to stabilize these drugs through encapsulation, storage, and in vivo release, and provide perspective on the most promising opportunities for the clinical translation of controlled-release peptides and proteins.
Collapse
Affiliation(s)
- Miusi Shi
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, Houston, TX 77030, USA; Department of Chemistry, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Büyükbayraktar HK, Pelit Arayıcı P, Ihlamur M, Gökkaya D, Karahan M, Abamor EŞ, Topuzoğulları M. Effect of polycation coating on the long-term pulsatile release of antigenic ESAT-6 1-20 peptide from PLGA nanoparticles. Colloids Surf B Biointerfaces 2023; 228:113421. [PMID: 37356137 DOI: 10.1016/j.colsurfb.2023.113421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/12/2023] [Accepted: 06/18/2023] [Indexed: 06/27/2023]
Abstract
The development of novel vaccine formulations against tuberculosis is necessary to reduce the number of new cases worldwide. Polymeric nanoparticles offer great potential as antigen delivery and immunostimulant systems for such purposes. In the study, we have encapsulated the antigenic peptide epitope of ESAT-6 protein of M. tuberculosis into PLGA nanoparticles and coated these nanoparticles with the cationic polymer of quaternized poly(4-vinylpyridine) (QPVP) to obtain a positively charged system as a potential nasal vaccine prototype. The produced spherical nanoparticles had hydrodynamic diameters between 180 and 240 nm with a narrow size distribution. The non-coated nanoparticle exhibited a 3-phase in vitro release profile that was completed in more than 4 months. In this release study, 5% of the peptide was released in the first 6 h and the nanoparticle remained silent until the 70th day. Then, an additional 5% of the peptide was released in 45 days. After coating the nanoparticle with QPVP, the release periods and peptide amounts dramatically changed. The antigenic peptide-loaded nanoparticles coated with the polycation stimulated the macrophages in vitro to release more nitric oxide (NO) compared to the free peptide and non-coated nanoparticle, which reveals the immunostimulant activity of the produced nanoparticle systems. The produced non-coated nanoparticles with the prolonged pulsatile release of the antigenic peptide can be used in the development of single injection self-boosting vaccine formulations. By coating these nanoparticles, both the release profile and immunogenicity can be changed.
Collapse
Affiliation(s)
- Hatice Kübra Büyükbayraktar
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, İstanbul, Turkiye
| | - Pelin Pelit Arayıcı
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, İstanbul, Turkiye
| | - Murat Ihlamur
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, İstanbul, Turkiye
| | - Damla Gökkaya
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, İstanbul, Turkiye
| | - Mesut Karahan
- Vocational School of Health Sciences Services, Üsküdar University, İstanbul, Turkiye
| | - Emrah Şefik Abamor
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, İstanbul, Turkiye
| | - Murat Topuzoğulları
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, İstanbul, Turkiye.
| |
Collapse
|
33
|
Heng WT, Lim HX, Tan KO, Poh CL. Validation of Multi-epitope Peptides Encapsulated in PLGA Nanoparticles Against Influenza A Virus. Pharm Res 2023; 40:1999-2025. [PMID: 37344603 DOI: 10.1007/s11095-023-03540-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND Influenza is a highly contagious respiratory disease which poses a serious threat to public health globally, causing severe diseases in 3-5 million humans and resulting in 650,000 deaths annually. The current licensed seasonal influenza vaccines lacked cross-reactivity against novel emerging influenza strains as they conferred limited neutralising capabilities. To address the issue, we designed a multi-epitope peptide-based vaccine delivered by the self-adjuvanting PLGA nanoparticles against influenza infections. METHODS A total of six conserved peptides representing B- and T-cell epitopes of Influenza A were identified and they were formulated in either incomplete Freund's adjuvant containing CpG ODN 1826 or being encapsulated in PLGA nanoparticles for the evaluation of immunogenicity in BALB/c mice. RESULTS The self-adjuvanting PLGA nanoparticles encapsulating the six conserved peptides were capable of eliciting the highest levels of IgG and IFN- γ producing cells. In addition, the immunogenicity of the six peptides encapsulated in PLGA nanoparticles showed greater humoral and cellular mediated immune responses elicited by the mixture of six naked peptides formulated in incomplete Freund's adjuvant containing CpG ODN 1826 in the immunized mice. Peptide 3 from the mixture of six peptides was found to exert necrotic effect on CD3+ T-cells and this finding indicated that peptide 3 should be removed from the nanovaccine formulation. CONCLUSION The study demonstrated the self-adjuvanting properties of the PLGA nanoparticles as a delivery system without the need for incorporation of toxic and costly conventional adjuvants in multi-epitope peptide-based vaccines.
Collapse
Affiliation(s)
- Wen Tzuen Heng
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Hui Xuan Lim
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Kuan Onn Tan
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), School of Medical and Life Sciences, Sunway University, No.5 Jalan Universiti, 47500, Petaling Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
34
|
Sun HC, Deng PM, Fu Y, Deng JH, Xie RH, Huang J, Qi M, Shi TY. Protective efficacy of Toxoplasma gondii GRA12 or GRA7 recombinant proteins encapsulated in PLGA nanoparticles against acute Toxoplasma gondii infection in mice. Front Cell Infect Microbiol 2023; 13:1209755. [PMID: 37502604 PMCID: PMC10368986 DOI: 10.3389/fcimb.2023.1209755] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
Background Toxoplasma gondii is an apicomplexan parasite that affects the health of humans and livestock, and an effective vaccine is urgently required. Nanoparticles can modulate and improve cellular and humoral immune responses. Methods In the current study, poly (D, L-lactic-co-glycolic acid) (PLGA) nanoparticles were used as a delivery system for the T. gondii dense granule antigens GRA12 and GRA7. BALB/c mice were injected with the vaccines and protective efficacy was evaluated. Results Mice immunized with PLGA+GRA12 exhibited significantly higher IgG, and a noticeable predominance of IgG2a over IgG1 was also observed. There was a 1.5-fold higher level of lymphocyte proliferation in PLGA+GRA12-injected mice compared to Alum+GRA12-immunized mice. Higher levels of IFN-g and IL-10 and a lower level of IL-4 were detected, indicating that Th1 and Th2 immune responses were induced but the predominant response was Th1. There were no significant differences between Alum+GRA7-immunized and PLGA+GRA7-immunized groups. Immunization with these four vaccines resulted in significantly reduced parasite loads, but they were lowest in PLGA+GRA12-immunized mice. The survival times of mice immunized with PLGA+GRA12 were also significantly longer than those of mice in the other vaccinated groups. Conclusion The current study indicated that T. gondii GRA12 recombinant protein encapsulated in PLGA nanoparticles is a promising vaccine against acute toxoplasmosis, but PLGA is almost useless for enhancing the immune response induced by T. gondii GRA7 recombinant protein.
Collapse
Affiliation(s)
- Hong-chao Sun
- Institute of Animal Husbandry and Veterinary Medicine, Department of Animal Parasitology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Pu-ming Deng
- Institute of Animal Science and Technology, Department of Animal Diseases Diagnosis and Control of Xinjiang Production & Construction Corps, Tarim University, Alar, China
| | - Yuan Fu
- Institute of Animal Husbandry and Veterinary Medicine, Department of Animal Parasitology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Jin-hua Deng
- Institute of Animal Science and Technology, Department of Animal Diseases Diagnosis and Control of Xinjiang Production & Construction Corps, Tarim University, Alar, China
| | - Rong-hui Xie
- Department of Animal Epidemic Surveillance, Zhejiang Provincial Animal Disease Prevention and Control Center, Hangzhou, China
| | - Jing Huang
- Department of Animal Epidemic Surveillance, Zhejiang Provincial Animal Disease Prevention and Control Center, Hangzhou, China
| | - Meng Qi
- Institute of Animal Science and Technology, Department of Animal Diseases Diagnosis and Control of Xinjiang Production & Construction Corps, Tarim University, Alar, China
| | - Tuan-yuan Shi
- Institute of Animal Husbandry and Veterinary Medicine, Department of Animal Parasitology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| |
Collapse
|
35
|
Li M, Pan Y, Xi Y, Wang M, Zeng Q. Insights and progress on epidemic characteristics, genotyping, and preventive measures of PEDV in China: A review. Microb Pathog 2023; 181:106185. [PMID: 37269880 DOI: 10.1016/j.micpath.2023.106185] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
Porcine Epidemic Diarrhoea (PED) is an acute, extremely infectious intestinal disease of pigs caused by the Porcine Epidemic Diarrhoea Virus (PEDV). The virus can affect pigs of all breeds and age groups and shows varying degrees of symptoms, with piglets, in particular, being infected with mortality rates of up to 100%. PEDV was first identified in China in the 1980s and in October 2010 a large-scale PED outbreak caused by a variant of PEDV occurred in China, resulting in huge economic losses. Initially, vaccination can effectively prevent the classical strain, but since December 2010, the PEDV variant has caused "persistent diarrhoea" with severe vomiting, watery diarrhoea, and high morbidity and mortality in newborn piglets as the dominant clinical features, with a significant increase in morbidity and mortality. This indicates that PEDV strains have mutated during evolution and that traditional vaccines no longer provide effective cross-immune protection, so it is necessary to optimize immunization programs and find effective treatments through epidemiological surveys of PEDV to reduce the economic losses caused by infections with mutated strains. This article reviews the progress of research on the aetiology, epidemiological characteristics, genotyping, pathogenesis, transmission routes, and comprehensive control of PEDV infection in China.
Collapse
Affiliation(s)
- Mei Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, Gansu, China
| | - Yao Xi
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China.
| | - Qiaoying Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China.
| |
Collapse
|
36
|
Alizadeh MH, Pooresmaeil M, Namazi H. Carboxymethyl cellulose@multi wall carbon nanotubes functionalized with Ugi reaction as a new curcumin carrier. Int J Biol Macromol 2023; 234:123778. [PMID: 36822289 DOI: 10.1016/j.ijbiomac.2023.123778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023]
Abstract
In recent years, the fabrication of new drug delivery systems (DDSs) based on functionalization by multi-component reactions (MCRs) has received special attention. In this regard, to obtain a new oral administration system for colon-specific cancer treatment, the CMC@MWCNTs@FCA carrier was designed and prepared from the functionalization of the CMC@MWCNTs as a biocompatible raw material with carboxamide group by the Ugi reaction. FT-IR analysis confirmed the successful synthesis of the product through the change in the functional groups of reagents. Additionally, the crystalline structure and porosity of the samples were studied by XRD and BET techniques. After a detailed characterization, the curcumin (CUR) was loaded on CMC@MWCNTs and CMC@MWCNTs@FCA, respectively, about 29 % and 38 %. In vitro drug release behavior studies for CUR-loaded CMC@MWCNTs@FCA showed the controlled release for it, so 11.6 % and 76.5 % of CUR, respectively were released at pH 1.2 and pH 7.4. Toxicological analysis displayed the IC50 of CMC@MWCNTs@FCA@CUR is 752 μg/mL. In conclusion, the obtained findings display that the fabricated system can be proposed as a biocompatible carrier for specific colon cancer treatment.
Collapse
Affiliation(s)
- Mohammad Hossein Alizadeh
- Polymer Research Laboratory, Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Malihe Pooresmaeil
- Polymer Research Laboratory, Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Hassan Namazi
- Polymer Research Laboratory, Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
37
|
Elzallat M, Hassan M, Elkramani N, Aboushousha T, AbdelLatif A, Helal N, Abu-Taleb H, El-Ahwany E. Nanoconjugated long non-coding RNA MEG3 as a new therapeutic approach for Hepatocellular carcinoma. Heliyon 2023; 9:e15288. [PMID: 37101621 PMCID: PMC10123146 DOI: 10.1016/j.heliyon.2023.e15288] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is an aggressive human cancer with a poor prognosis. Long non-coding RNAs (lncRNA) have multiple functions: epigenomic regulation, gene transcription, protein-coding gene translation, and genome defense. The involvement of lncRNAs in therapy offers a vast step in cancer treatment. Objective In the current study, a novel therapeutic regimen using polymer nanoparticle-mediated delivery of lncRNA was designed to control the progression of hepatocarcinogenesis. Methods One hundred mice were divided into 5 groups. The first group served as a normal-control group and was injected with saline, whereas the pathological-control group (the second group) was injected with N-Nitrosodiethylamine (DEN) weekly for 16 weeks. Group 3, Group 4, and Group 5 were injected intrahepatically with polymer nanoparticles (NPs) alone, lncRNA MEG3 alone, and conjugated NPs, respectively, once/week for four weeks starting on the 12th week after DEN injection. After 16 weeks, animals were euthanized, and liver specimens and blood samples were collected for pathological, molecular, and biochemical assessment. Results Compared to the pathological-control group, nanoconjugates lncRNA MEG3 demonstrated a significant improvement in histopathology and tumour-associated biomarkers. Furthermore, the expression of the SENP1 and PCNA was downregulated. Conclusion MEG3 conjugated nanoparticles can be considered a novel therapeutic regimen for HCC.
Collapse
|
38
|
马 龙, 罗 向, 莫 丽, 樊 家, 刘 大. [Mechanism of Der f 1/IGF-1 nanoparticle promoting the production of regulatory T cell]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2023; 37:272-277. [PMID: 36987957 PMCID: PMC10406592 DOI: 10.13201/j.issn.2096-7993.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Indexed: 03/30/2023]
Abstract
Objective:To prepare PLGA nanoparticles loaded with Der f 1/IGF-1(Der f 1/IGF-1 NPs) and investigate their role in promoting the formation of Treg cells. Methods:NPs coated with Der f 1/IGF-1 were prepared by double emulsion method and their physicochemical properties and cumulative release rate in vitro were analyzed. After pretreatment, BMDC was divided into Saline group, Blank NPs group, Der f 1/IGF-1 group and Der f 1/IGF-1 NPs group. Determination of the expression of IL-10 and TGF-β in BMDC by ELISA. The number of Treg cells was detected by flow cytometry. Results:The results showed that Der f 1/IGF-1 NPs were spherical structures, with good dispersion, particle size less than 200 nm, negative charge and stable slow-release effect of Zeta potential. After BMDC pretreatment, the expression levels of TGF-β and IL-10 in BMDC cells in the Der f 1/IGF-1 NPs group were significantly increased compared with the Blank NPs group, and the difference was statistically significant(P<0.001). After co-culture with CD4+ T cells, the proportion of Treg cells produced in the Der f 1/IGF-1 NPs group was significantly increased, and the difference was statistically significant(P<0.001). Conclusion:Der f 1/IGF-1 NPs can induce Treg cell generation in vitro. This study provides a new and more effective method for the reconstruction of immune tolerance dysfunction.
Collapse
Affiliation(s)
- 龙鹏 马
- 南方医科大学深圳医院儿童耳鼻咽喉科(广东深圳,518060)Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518060, China
| | - 向前 罗
- 南方医科大学深圳医院儿童耳鼻咽喉科(广东深圳,518060)Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518060, China
| | - 丽华 莫
- 南方医科大学深圳医院儿童耳鼻咽喉科(广东深圳,518060)Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518060, China
| | - 家良 樊
- 南方医科大学深圳医院儿童耳鼻咽喉科(广东深圳,518060)Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518060, China
| | - 大波 刘
- 南方医科大学深圳医院儿童耳鼻咽喉科(广东深圳,518060)Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518060, China
| |
Collapse
|
39
|
Qu Y, De Rose R, Kim C, Zhou J, Lin Z, Ju Y, Bhangu SK, Cortez‐Jugo C, Cavalieri F, Caruso F. Supramolecular Polyphenol-DNA Microparticles for In Vivo Adjuvant and Antigen Co-Delivery and Immune Stimulation. Angew Chem Int Ed Engl 2023; 62:e202214935. [PMID: 36700351 PMCID: PMC10946467 DOI: 10.1002/anie.202214935] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
DNA-based materials have attracted interest due to the tunable structure and encoded biological functionality of nucleic acids. A simple and general approach to synthesize DNA-based materials with fine control over morphology and bioactivity is important to expand their applications. Here, we report the synthesis of DNA-based particles via the supramolecular assembly of tannic acid (TA) and DNA. Uniform particles with different morphologies are obtained using a variety of DNA building blocks. The particles enable the co-delivery of cytosine-guanine adjuvant sequences and the antigen ovalbumin in model cells. Intramuscular injection of the particles in mice induces antigen-specific antibody production and T cell responses with no apparent toxicity. Protein expression in cells is shown using capsules assembled from TA and plasmid DNA. This work highlights the potential of TA as a universal material for directing the supramolecular assembly of DNA into gene and vaccine delivery platforms.
Collapse
Affiliation(s)
- Yijiao Qu
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Robert De Rose
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Chan‐Jin Kim
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Jiajing Zhou
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Zhixing Lin
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Yi Ju
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Sukhvir Kaur Bhangu
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Christina Cortez‐Jugo
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie Chimiche Universita' di Roma “Tor Vergata”Via della Ricerca Scientifica 100133RomeItaly
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| |
Collapse
|
40
|
Kumar BA, Panickan S, Bindu S, Kumar V, Ramakrishnan S, Sonal, Shrivastava S, Dandapat S. Immunogenicity and protective efficacy of an inactivated Newcastle disease virus vaccine encapsulated in poly-(lactic-co-glycolic acid) nanoparticles. Poult Sci 2023; 102:102679. [PMID: 37116285 PMCID: PMC10160591 DOI: 10.1016/j.psj.2023.102679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
An immunization experiment was conducted in specific pathogen-free chickens with the inactivated Newcastle disease virus (NDV) vaccine encapsulated in the poly-(lactic-co-glycolic) acid (PLGA) nanoparticles (NP) to evaluate its immunogenicity and protective efficacy. The NDV vaccine was prepared by inactivating one virulent Indian strain of NDV belonging to Genotype VII by using beta-propiolactone. PLGA nanoparticles encapsulating inactivated NDV were prepared by the solvent evaporation method. Scanning electron microscopy and zeta sizer analysis revealed that the (PLGA+NDV) NP were spherical, with an average size of 300 nm, having a zeta potential of -6 mV. The encapsulation efficiency and loading efficiency were 72% and 2.4%, respectively. On immunization trial in chicken, the (PLGA+NDV) NP induced significantly (P < 0.0001) higher levels of HI and IgY antibodies with the peak HI titer of 28 and higher expression of IL-4 mRNA. The consistency of higher antibody levels suggests slow and pulsatile release of the antigens from the (PLGA+NDV) NP. The nano-NDV vaccine also induced cell mediated immunity with higher expression of IFN-γ indicating strong Th1 mediated immune responses in contrast to the commercial oil adjuvanted inactivated NDV vaccine. Further, the (PLGA+NDV) NP afforded 100% protection against the virulent NDV challenge. Our results suggested that PLGA NP have adjuvant potential on induction of humoral as well as Th1 biased cell mediated immune responses and also enhanced protective efficacy of the inactivated NDV vaccine. This study provides an insight for development of PLGA NP based inactivated NDV vaccine using the same genotype circulating in the field as well as for other avian diseases at exigencies.
Collapse
|
41
|
Umar AK. Stem Cell's Secretome Delivery Systems. Adv Pharm Bull 2023; 13:244-258. [PMID: 37342369 PMCID: PMC10278206 DOI: 10.34172/apb.2023.027] [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: 04/17/2021] [Revised: 10/05/2021] [Accepted: 12/31/2021] [Indexed: 09/01/2023] Open
Abstract
Stem cells' secretome contains biomolecules that are ready to give therapeutic activities. However, the biomolecules should not be administered directly because of their in vivo instability. They can be degraded by enzymes or seep into other tissues. There have been some advancements in localized and stabilized secretome delivery systems, which have increased their effectiveness. Fibrous, in situ, or viscoelastic hydrogel, sponge-scaffold, bead powder/ suspension, and bio-mimetic coating can maintain secretome retention in the target tissue and prolong the therapy by sustained release. Porosity, young's modulus, surface charge, interfacial interaction, particle size, adhesiveness, water absorption ability, in situ gel/film, and viscoelasticity of the preparation significantly affect the quality, quantity, and efficacy of the secretome. Therefore, the dosage forms, base materials, and characteristics of each system need to be examined to develop a more optimal secretome delivery system. This article discusses the clinical obstacles and potential solutions for secretome delivery, characterization of delivery systems, and devices used or potentially used in secretome delivery for therapeutic applications. This article concludes that secretome delivery for various organ therapies necessitates the use of different delivery systems and bases. Coating, muco-, and cell-adhesive systems are required for systemic delivery and to prevent metabolism. The lyophilized form is required for inhalational delivery, and the lipophilic system can deliver secretomes across the blood-brain barrier. Nano-sized encapsulation and surface-modified systems can deliver secretome to the liver and kidney. These dosage forms can be administered using devices such as a sprayer, eye drop, inhaler, syringe, and implant to improve their efficacy through dosing, direct delivery to target tissues, preserving stability and sterility, and reducing the immune response.
Collapse
Affiliation(s)
- Abd. Kakhar Umar
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| |
Collapse
|
42
|
Peletta A, Lemoine C, Courant T, Collin N, Borchard G. Meeting vaccine formulation challenges in an emergency setting: Towards the development of accessible vaccines. Pharmacol Res 2023; 189:106699. [PMID: 36796463 DOI: 10.1016/j.phrs.2023.106699] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Vaccination is considered one of the most successful strategies to prevent infectious diseases. In the event of a pandemic or epidemic, the rapid development and distribution of the vaccine to the population is essential to reduce mortality, morbidity and transmission. As seen during the COVID-19 pandemic, the production and distribution of vaccines has been challenging, in particular for resource-constrained settings, essentially slowing down the process of achieving global coverage. Pricing, storage, transportation and delivery requirements of several vaccines developed in high-income countries resulted in limited access for low-and-middle income countries (LMICs). The capacity to manufacture vaccines locally would greatly improve global vaccine access. In particular, for the development of classical subunit vaccines, the access to vaccine adjuvants is a pre-requisite for more equitable access to vaccines. Vaccine adjuvants are agents required to augment or potentiate, and possibly target the specific immune response to such type of vaccine antigens. Openly accessible or locally produced vaccine adjuvants may allow for faster immunization of the global population. For local research and development of adjuvanted vaccines to expand, knowledge on vaccine formulation is of paramount importance. In this review, we aim to discuss the optimal characteristics of a vaccine developed in an emergency setting by focusing on the importance of vaccine formulation, appropriate use of adjuvants and how this may help overcome barriers for vaccine development and production in LMICs, achieve improved vaccine regimens, delivery and storage requirements.
Collapse
Affiliation(s)
- Allegra Peletta
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| | - Céline Lemoine
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Thomas Courant
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| |
Collapse
|
43
|
Nooraei S, Sarkar Lotfabadi A, Akbarzadehmoallemkolaei M, Rezaei N. Immunogenicity of Different Types of Adjuvants and Nano-Adjuvants in Veterinary Vaccines: A Comprehensive Review. Vaccines (Basel) 2023; 11:vaccines11020453. [PMID: 36851331 PMCID: PMC9962389 DOI: 10.3390/vaccines11020453] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Vaccination is the best way to prevent and reduce the damage caused by infectious diseases in animals and humans. So, several vaccines are used for prophylactic purposes before the pathogen infects, while therapeutic vaccines strengthen the immune system after infection with the pathogen. Adjuvants are molecules, compounds, or macromolecules that enhance non-specific immunity and, in collaboration with antigen(s), can improve the body's immune responses and change the type of immune response. The potential and toxicity of adjuvants must be balanced to provide the safest stimulation with the fewest side effects. In order to overcome the limitations of adjuvants and the effective and controlled delivery of antigens, attention has been drawn to nano-carriers that can be a promising platform for better presenting and stimulating the immune system. Some studies show that nanoparticles have a more remarkable ability to act as adjuvants than microparticles. Because nano-adjuvants inactively target antigen-presenting cells (APCs) and change their chemical surface, nanoparticles also perform better in targeted antigen delivery because they cross biological barriers more easily. We collected and reviewed various types of nano-adjuvants with their specific roles in immunogenicity as a prominent strategy used in veterinary vaccines in this paper.
Collapse
Affiliation(s)
- Soren Nooraei
- Faculty of Veterinary Medicine, Shahrekord University, Shahrekord 8818634141, Iran
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Alireza Sarkar Lotfabadi
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Milad Akbarzadehmoallemkolaei
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Nima Rezaei
- Animal Model Integrated Network (AMIN), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran 1419733151, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417653761, Iran
- Correspondence:
| |
Collapse
|
44
|
Horvath D, Basler M. PLGA Particles in Immunotherapy. Pharmaceutics 2023; 15:pharmaceutics15020615. [PMID: 36839937 PMCID: PMC9965784 DOI: 10.3390/pharmaceutics15020615] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Poly(lactic-co-glycolic acid) (PLGA) particles are a widely used and extensively studied drug delivery system. The favorable properties of PLGA such as good bioavailability, controlled release, and an excellent safety profile due to the biodegradable polymer backbone qualified PLGA particles for approval by the authorities for the application as a drug delivery platform in humas. In recent years, immunotherapy has been established as a potent treatment option for a variety of diseases. However, immunomodulating drugs rely on targeted delivery to specific immune cell subsets and are often rapidly eliminated from the system. Loading of PLGA particles with drugs for immunotherapy can protect the therapeutic compounds from premature degradation, direct the drug delivery to specific tissues or cells, and ensure sustained and controlled drug release. These properties present PLGA particles as an ideal platform for immunotherapy. Here, we review recent advances of particulate PLGA delivery systems in the application for immunotherapy in the fields of allergy, autoimmunity, infectious diseases, and cancer.
Collapse
Affiliation(s)
- Dennis Horvath
- Division of Immunology, Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, D-78457 Konstanz, Germany
| | - Michael Basler
- Division of Immunology, Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, CH-8280 Kreuzlingen, Switzerland
- Correspondence:
| |
Collapse
|
45
|
Patil S, Vijayanand S, Joshi D, Menon I, Braz Gomes K, Kale A, Bagwe P, Yacoub S, Uddin MN, D'Souza MJ. Subunit microparticulate vaccine delivery using microneedles trigger significant SARS-spike-specific humoral and cellular responses in a preclinical murine model. Int J Pharm 2023; 632:122583. [PMID: 36610521 PMCID: PMC9811858 DOI: 10.1016/j.ijpharm.2023.122583] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
The objective of this "proof-of-concept" study was to evaluate the synergistic effect of a subunit microparticulate vaccine and microneedles (MN) assisted vaccine delivery system against a human coronavirus. Here, we formulated PLGA polymeric microparticles (MPs) encapsulating spike glycoprotein (GP) of SARS-CoV as the model antigen. Similarly, we formulated adjuvant MPs encapsulating Alhydrogel® and AddaVax™. The antigen/adjuvant MPs were characterized and tested in vitro for immunogenicity. We found that the antigen/adjuvant MPs were non-cytotoxic in vitro. The spike GP MPs + Alhydrogel® MPs + AddaVax™ MPs showed enhanced immunogenicity in vitro as confirmed through the release of nitrite, autophagy, and antigen presenting molecules with their co-stimulatory molecules. Next, we tested the in vivo efficacy of the spike GP MP vaccine with and without adjuvant MPs in mice vaccinated using MN. The spike GP MPs + Alhydrogel® MPs + AddaVax™ MPs induced heightened spike GP-specific IgG, IgG1 and IgG2a antibodies in mice. Also, spike GP MPs + Alhydrogel® MPs + AddaVax™ MPs enhanced expression of CD4+ and CD8+ T cells in secondary lymphoid organ like spleen. These results indicated spike GP-specific humoral immunity and cellular immunity in vivo. Thus, we employed the benefits of both the subunit vaccine MPs and dissolving MN to form a non-invasive and effective vaccination strategy against human coronaviruses.
Collapse
Affiliation(s)
- Smital Patil
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Sharon Vijayanand
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Devyani Joshi
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Ipshita Menon
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Keegan Braz Gomes
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Akanksha Kale
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Priyal Bagwe
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Shadi Yacoub
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Mohammad N Uddin
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Martin J D'Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA.
| |
Collapse
|
46
|
Batty CJ, Lifshits LM, Hendy DA, Eckshtain-Levi M, Ontiveros-Padilla LA, Carlock MA, Ross TM, Bachelder EM, Ainslie KM. Vinyl Sulfone-functionalized Acetalated Dextran Microparticles as a Subunit Broadly Acting Influenza Vaccine. AAPS J 2023; 25:22. [PMID: 36720729 DOI: 10.1208/s12248-023-00786-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 01/18/2023] [Indexed: 02/02/2023] Open
Abstract
Influenza is a global health concern with millions of infections occurring yearly. Seasonal flu vaccines are one way to combat this virus; however, they are poorly protective against influenza as the virus is constantly mutating, particularly at the immunodominant hemagglutinin (HA) head group. A more broadly acting approach involves Computationally Optimized Broadly Reactive Antigen (COBRA). COBRA HA generates a broad immune response that is capable of protecting against mutating strains. Unfortunately, protein-based vaccines are often weekly immunogenic, so to help boost the immune response, we employed the use of acetalated dextran (Ace-DEX) microparticles (MPs) two ways: one to conjugate COBRA HA to the surface and a second to encapsulate cGAMP. To conjugate the COBRA HA to the surface of the Ace-DEX MPs, a poly(L-lactide)-polyethylene glycol co-polymer with a vinyl sulfone terminal group (PLLA-PEG-VS) was used. MPs encapsulating the STING agonist cGAMP were co-delivered with the antigen to form a broadly active influenza vaccine. This vaccine approach was evaluated in vivo with a prime-boost-boost vaccination schedule and illustrated generation of a humoral and cellular response that could protect against a lethal challenge of A/California/07/2009 in BALB/c mice.
Collapse
Affiliation(s)
- Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Liubov M Lifshits
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Meital Eckshtain-Levi
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Luis A Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Michael A Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, Florida, USA
| | - Ted M Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, Florida, USA.,Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA.,Department of Infectious Diseases, University of Georgia, Athens, Goergia, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4211 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA. .,Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA. .,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, USA.
| |
Collapse
|
47
|
Muñoz-Wolf N, Ward RW, Hearnden CH, Sharp FA, Geoghegan J, O’Grady K, McEntee CP, Shanahan KA, Guy C, Bowie AG, Campbell M, Roces C, Anderluzzi G, Webb C, Perrie Y, Creagh E, Lavelle EC. Non-canonical inflammasome activation mediates the adjuvanticity of nanoparticles. Cell Rep Med 2023; 4:100899. [PMID: 36652908 PMCID: PMC9873954 DOI: 10.1016/j.xcrm.2022.100899] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/24/2022] [Accepted: 12/19/2022] [Indexed: 01/19/2023]
Abstract
The non-canonical inflammasome sensor caspase-11 and gasdermin D (GSDMD) drive inflammation and pyroptosis, a type of immunogenic cell death that favors cell-mediated immunity (CMI) in cancer, infection, and autoimmunity. Here we show that caspase-11 and GSDMD are required for CD8+ and Th1 responses induced by nanoparticulate vaccine adjuvants. We demonstrate that nanoparticle-induced reactive oxygen species (ROS) are size dependent and essential for CMI, and we identify 50- to 60-nm nanoparticles as optimal inducers of ROS, GSDMD activation, and Th1 and CD8+ responses. We reveal a division of labor for IL-1 and IL-18, where IL-1 supports Th1 and IL-18 promotes CD8+ responses. Exploiting size as a key attribute, we demonstrate that biodegradable poly-lactic co-glycolic acid nanoparticles are potent CMI-inducing adjuvants. Our work implicates ROS and the non-canonical inflammasome in the mode of action of polymeric nanoparticulate adjuvants and establishes adjuvant size as a key design principle for vaccines against cancer and intracellular pathogens.
Collapse
Affiliation(s)
- Natalia Muñoz-Wolf
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland,Translational & Respiratory Immunology Lab, Department of Clinical Medicine, School of Medicine, Trinity Biomedical Sciences Institute, Dublin D02 R590, Ireland,Clinical Medicine Tallaght University Hospital, Dublin D24 NR04, Ireland
| | - Ross W. Ward
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Claire H. Hearnden
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Fiona A. Sharp
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Joan Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland,Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Katie O’Grady
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Craig P. McEntee
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Katharine A. Shanahan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Coralie Guy
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Andrew G. Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Carla.B. Roces
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Giulia Anderluzzi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Cameron Webb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Emma Creagh
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Ed C. Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin D02 PN40, Ireland,Corresponding author
| |
Collapse
|
48
|
Jamaledin R, Sartorius R, Di Natale C, Onesto V, Manco R, Mollo V, Vecchione R, De Berardinis P, Netti PA. PLGA microparticle formulations for tunable delivery of a nano-engineered filamentous bacteriophage-based vaccine: in vitro and in silico-supported approach. JOURNAL OF NANOSTRUCTURE IN CHEMISTRY 2023:1-16. [PMID: 36687278 PMCID: PMC9838389 DOI: 10.1007/s40097-022-00519-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 06/17/2023]
Abstract
Bacteriophages have attracted great attention in the bioengineering field in diverse research areas from tissue engineering to therapeutic and clinical applications. Recombinant filamentous bacteriophage, carrying multiple copies of foreign peptides on protein capsid has been successfully used in the vaccine delivery setting, even if their plasma instability and degradation have limited their use on the pharmaceutical market. Encapsulation techniques in polymeric materials can be applied to preserve bacteriophage activity, extend its half-life, and finely regulate their release in the target environment. The main goal of this study was to provide tunable formulations of the bacteriophage encapsulated in polymeric microparticles (MPs). We used poly (lactic-co-glycolic-acid) as a biocompatible and biodegradable polymer with ammonium bicarbonate as a porogen to encapsulate bacteriophage expressing OVA (257-264) antigenic peptide. We demonstrate that nano-engineered fdOVA bacteriophages encapsulated in MPs preserve their structure and are immunologically active, inducing a strong immune response towards the delivered peptide. Moreover, MP encapsulation prolongs bacteriophage stability over time also at room temperature. Additionally, in this study, we show the ability of in silico-supported approach to predict and tune the release of bacteriophages. These results lay the framework for a versatile bacteriophage-based vaccine delivery system that could successfully generate robust immune responses in a sustained manner, to be used as a platform against cancer and new emerging diseases. Graphical abstract Synopsis: administration of recombinant bacteriophage-loaded PLGA microparticles for antigen delivery. PLGA microparticles release the bacteriophages, inducing activation of dendritic cells and enhancing antigen presentation and specific T cell response. Bacteriophage-encapsulated microneedles potentially can be administered into human body and generate robust immune responses.
Collapse
Affiliation(s)
- Rezvan Jamaledin
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King’s Buildings, Edinburgh, EH9 3JL, UK
| | - Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 80131 Naples, Italy
| | - Concetta Di Natale
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre On Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy
| | - Valentina Onesto
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy
| | - Roberta Manco
- Institute of Biochemistry and Cell Biology (IBBC), CNR, 80131 Naples, Italy
| | - Valentina Mollo
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy
| | - Raffaele Vecchione
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy
| | | | - Paolo Antonio Netti
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre On Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
- Department of Chemical Materials and Industrial Production (DICMAPI), University of Naples Federico II, Naples, Italy
| |
Collapse
|
49
|
Ho E, Deng Y, Akbar D, Da K, Létourneau M, Morshead CM, Chatenet D, Shoichet MS. Tunable Surface Charge Enables the Electrostatic Adsorption-Controlled Release of Neuroprotective Peptides from a Hydrogel-Nanoparticle Drug Delivery System. ACS APPLIED MATERIALS & INTERFACES 2023; 15:91-105. [PMID: 36520607 DOI: 10.1021/acsami.2c17631] [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: 06/17/2023]
Abstract
We exploit the electrostatic interactions between the positively charged neuroprotective peptide, pituitary adenylate cyclase-activating polypeptide (PACAP), and negatively charged poly(lactic-co-glycolic acid) (PLGA) nanoparticles to control PACAP release from the surface of nanoparticles dispersed in a hyaluronan-methylcellulose (HAMC) hydrogel composite. PACAP is a promising therapeutic for the treatment of neurological disorders, yet it has been difficult to deliver in vivo. Herein, the PACAP release rate was tuned by manipulating peptide adsorption onto the surface of blank nanoparticles by modifying either nanoparticle loading in the hydrogel or nanoparticle surface charge. This peptide-nanoparticle interaction was controlled by the pH-responsive carboxylic acid end terminal groups of PLGA. We further validated this system with the controlled release of a novel stabilized PACAP analog: Ac-[Ala15, Ala20]PACAP-propylamide, which masks its recognition to peptidases in circulation. Both wild-type and stabilized PACAP released from the vehicle increased the production of neuroprotective Interleukin-6 from cultured primary astrocytes. Using computational fluid dynamics methods, PACAP release from the composite was predicted based on experimentally derived adsorption isotherms, which exhibited similar release profiles to experimental data. This versatile adsorption-based system was used to deliver PACAP locally to the brains of stroke-injured mice over a 10 day period in vivo, highlighting its effectiveness for the controlled release of PACAP to the central nervous system.
Collapse
Affiliation(s)
- Eric Ho
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, OntarioM5S 3G9, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| | - Yaoqi Deng
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| | - Dania Akbar
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| | - Kevin Da
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
| | - Myriam Létourneau
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QuebecH7 V 1B7, Canada
| | - Cindi M Morshead
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, OntarioM5S 3G9, Canada
- Department of Surgery, University of Toronto, 149 College Street, Toronto, OntarioM5S 3E1, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, OntarioM5S 3E1, Canada
| | - David Chatenet
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QuebecH7 V 1B7, Canada
| | - Molly S Shoichet
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, OntarioM5S 3G9, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, OntarioM5S 3E5, Canada
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, OntarioM5S 3E1, Canada
| |
Collapse
|
50
|
Liu M, Thijssen S, Hennink WE, Garssen J, van Nostrum CF, Willemsen LM. Oral pretreatment with β-lactoglobulin derived peptide and CpG co-encapsulated in PLGA nanoparticles prior to sensitizations attenuates cow's milk allergy development in mice. Front Immunol 2023; 13:1053107. [PMID: 36703973 PMCID: PMC9872660 DOI: 10.3389/fimmu.2022.1053107] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Cow's milk allergy is a common food allergy among infants. Improved hygiene conditions and loss of microbial diversity are associated with increased risk of allergy development. The intestinal immune system is essential for oral tolerance induction. In this respect, bacterial CpG DNA is known to drive Th1 and regulatory T-cell (Treg) development via Toll-Like-Receptor 9 (TLR-9) signaling, skewing away from the allergic Th2 phenotype. We aimed to induce allergen specific tolerance via oral delivery of poly (lactic-co-glycolic acid) nanoparticles (NP) co-encapsulated with a selected β-lactoglobulin derived peptide (BLG-Pep) and TLR-9 ligand CpG oligodeoxynucleotide (CpG). In vivo, 3-4-week-old female C3H/HeOuJ mice housed in individually ventilated cages received 6-consecutive-daily gavages of either PBS, whey, BLG-Pep/NP, CpG/NP, a mixture of BLG-Pep/NP plus CpG/NP or co-encapsulated BLG-Pep+CpG/NP, before 5-weekly oral sensitizations with whey plus cholera toxin (CT) or only CT (sham) and were challenged with whey 5 days after the last sensitization. The co-encapsulated BLG-Pep+CpG/NP pretreatment, but not BLG-Pep/NP, CpG/NP or the mixture of BLG-Pep/NP plus CpG/NP, prevented the whey-induced allergic skin reactivity and prevented rise in serum BLG-specific IgE compared to whey-sensitized mice. Importantly, co-encapsulated BLG-Pep+CpG/NP pretreatment reduced dendritic cell (DC) activation and lowered the frequencies of PD-L1+ DC in the mesenteric lymph nodes compared to whey-sensitized mice. By contrast, co-encapsulated BLG-Pep+CpG/NP pretreatment increased the frequency of splenic PD-L1+ DC compared to the BLG-Pep/NP plus CpG/NP recipients, in association with lower Th2 development and increased Treg/Th2 and Th1/Th2 ratios in the spleen. Oral administration of PLGA NP co-encapsulated with BLG-Pep and CpG prevented rise in serum BLG-specific IgE and symptom development while lowering splenic Th2 cell frequency in these mice which were kept under strict hygienic conditions.
Collapse
Affiliation(s)
- Mengshan Liu
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Suzan Thijssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Wim E. Hennink
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,Department of Immunology, Nutricia Research B.V., Utrecht, Netherlands
| | - Cornelus F. van Nostrum
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,*Correspondence: Linette E. M. Willemsen,
| |
Collapse
|