1
|
Daneshvar A, Farokhi M, Bonakdar S, Vossoughi M. Synthesis and characterization of injectable thermosensitive hydrogel based on Pluronic-grafted silk fibroin copolymer containing hydroxyapatite nanoparticles as potential for bone tissue engineering. Int J Biol Macromol 2024; 277:134412. [PMID: 39097043 DOI: 10.1016/j.ijbiomac.2024.134412] [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: 04/04/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Injectable hydrogels are promising for bone tissue engineering due to their minimally invasive application and adaptability to irregular defects. This study presents the development of pluronic grafted silk fibroin (PF-127-g-SF), a temperature-sensitive graft copolymer synthesized from SF and modified PF-127 via a carbodiimide coupling reaction. The PF-127-g-SF copolymer exhibited a higher sol-gel transition temperature (34 °C at 16 % w/v) compared to PF-127 (23 °C), making it suitable for injectable applications. It also showed improved flexibility and strength, with a yielding point increase from <10 % to nearly 30 %. Unlike PF-127 gel, which degrades within 72 h in aqueous media, the PF-127-g-SF copolymer maintained a stable gel structure for over two weeks due to its robust crosslinked hydrogel network. Incorporating hydroxyapatite nanoparticles (n-HA) into the hydrogel reduced pore size and decreased swelling and degradation rates, extending structural stability to four weeks. Increasing n-HA concentration from 0 % to 20 % reduced porosity from 80 % to 66 %. Rheological studies indicated that n-HA enhanced the scaffold's strength and mechanical properties without altering gelation temperature. Cellular studies with MG-63 cells showed that n-HA concentration influenced cell viability and mineralization, highlighting the scaffold's potential in bone tissue engineering.
Collapse
Affiliation(s)
- Anahita Daneshvar
- Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Sharif University of Technology, Tehran 14588-89694, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.
| | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Manouchehr Vossoughi
- Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Sharif University of Technology, Tehran 14588-89694, Iran; Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
2
|
Kunkel AA, McHugh KJ. Injectable controlled-release systems for the prevention and treatment of infectious diseases. J Biomed Mater Res A 2024; 112:1224-1240. [PMID: 37740704 DOI: 10.1002/jbm.a.37615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/25/2023]
Abstract
Pharmaceutical drugs, including vaccines, pre- and post-exposure prophylactics, and chronic drug therapies, are crucial tools in the prevention and treatment of infectious diseases. These drugs have the ability to increase survival and improve patient quality of life; however, infectious diseases still accounted for more than 10.2 million deaths in 2019 before the COVID-19 pandemic. High mortality can be, in part, attributed to challenges in the availability of adequate drugs and vaccines, limited accessibility, poor drug bioavailability, the high cost of some treatments, and low patient adherence. A majority of these factors are logistical rather than technical challenges, providing an opportunity for existing drugs and vaccines to be improved through formulation. Injectable controlled-release drug delivery systems are one class of formulations that have the potential to overcome many of these limitations by releasing their contents in a sustained manner to reduce the need for frequent re-administration and improve clinical outcomes. This review provides an overview of injectable controlled drug delivery platforms, including microparticles, nanoparticles, and injectable gels, detailing recent developments using these systems for single-injection vaccination, long-acting prophylaxis, and sustained-release treatments for infectious disease.
Collapse
Affiliation(s)
- Alyssa A Kunkel
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Department of Chemistry, Rice University, Houston, Texas, USA
| |
Collapse
|
3
|
Masimov R, Wasan EK. Chitosan non-particulate vaccine delivery systems. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12921. [PMID: 39114808 PMCID: PMC11303186 DOI: 10.3389/jpps.2024.12921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024]
Abstract
Chitosan is an extensively used polymer for drug delivery applications in particulate and non-particulate carriers. Chitosan-based particulate, nano-, and microparticle, carriers have been the most extensively studied for the delivery of therapeutics and vaccines. However, chitosan has also been used in vaccine applications for its adjuvant properties in various hydrogels or as a carrier coating material. The focus of this review will be on the usage of chitosan as a vaccine adjuvant based on its intrinsic immunogenicity; the various forms of chitosan-based non-particulate delivery systems such as thermosensitive hydrogels, microneedles, and conjugates; and the advantages of its role as a coating material for vaccine carriers.
Collapse
Affiliation(s)
| | - Ellen K. Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
4
|
He W, Zhang Y, Qu Y, Liu M, Li G, Pan L, Xu X, Shi G, Hao Q, Liu F, Gao Y. Research progress on hydrogel-based drug therapy in melanoma immunotherapy. BMB Rep 2024; 57:71-78. [PMID: 38053295 PMCID: PMC10910090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/04/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
Melanoma is one of the most aggressive skin tumors, and conventional treatment modalities are not effective in treating advanced melanoma. Although immunotherapy is an effective treatment for melanoma, it has disadvantages, such as a poor response rate and serious systemic immune-related toxic side effects. The main solution to this problem is the use of biological materials such as hydrogels to reduce these side effects and amplify the immune killing effect against tumor cells. Hydrogels have great advantages as local slow-release drug carriers, including the ability to deliver antitumor drugs directly to the tumor site, enhance the local drug concentration in tumor tissue, reduce systemic drug distribution and exhibit good degradability. Despite these advantages, there has been limited research on the application of hydrogels in melanoma treatment. Therefore, this article provides a comprehensive review of the potential application of hydrogels in melanoma immunotherapy. Hydrogels can serve as carriers for sustained drug delivery, enabling the targeted and localized delivery of drugs with minimal systemic side effects. This approach has the potential to improve the efficacy of immunotherapy for melanoma. Thus, the use of hydrogels as drug delivery vehicles for melanoma immunotherapy has great potential and warrants further exploration. [BMB Reports 2024; 57(2): 71-78].
Collapse
Affiliation(s)
- Wei He
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Yanqin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Air Force Military Medical University, Xi’an 710032, China
| | - Yi Qu
- Department of Xi’an Shunmei Medical Cosmetology Outpatient, Xi’an 710075, China
| | - Mengmeng Liu
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Guodong Li
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Luxiang Pan
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Xinyao Xu
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Gege Shi
- College of Life Science, Northwest University, Xi’an 710069, China
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Fen Liu
- Department of Periodontology, Shenzhen Stomatological Hospital (Pingshan), Southern Medical University, Shenzhen 510515, China
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
5
|
Miwa H, Antao OQ, Kelly‐Scumpia KM, Baghdasarian S, Mayer DP, Shang L, Sanchez GM, Archang MM, Scumpia PO, Weinstein JS, Di Carlo D. Improved Humoral Immunity and Protection against Influenza Virus Infection with a 3d Porous Biomaterial Vaccine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302248. [PMID: 37750461 PMCID: PMC10625058 DOI: 10.1002/advs.202302248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/03/2023] [Indexed: 09/27/2023]
Abstract
New vaccine platforms that activate humoral immunity and generate neutralizing antibodies are required to combat emerging pathogens, including influenza virus. A slurry of antigen-loaded hydrogel microparticles that anneal to form a porous scaffold with high surface area for antigen uptake by infiltrating immune cells as the biomaterial degrades is demonstrated to enhance humoral immunity. Antigen-loaded-microgels elicited a robust cellular humoral immune response, with increased CD4+ T follicular helper (Tfh) cells and prolonged germinal center (GC) B cells comparable to the commonly used adjuvant, aluminum hydroxide (Alum). Increasing the weight fraction of polymer material led to increased material stiffness and antigen-specific antibody titers superior to Alum. Vaccinating mice with inactivated influenza virus loaded into this more highly cross-linked formulation elicited a strong antibody response and provided protection against a high dose viral challenge. By tuning physical and chemical properties, adjuvanticity can be enhanced leading to humoral immunity and protection against a pathogen, leveraging two different types of antigenic material: individual protein antigen and inactivated virus. The flexibility of the platform may enable design of new vaccines to enhance innate and adaptive immune cell programming to generate and tune high affinity antibodies, a promising approach to generate long-lasting immunity.
Collapse
Affiliation(s)
- Hiromi Miwa
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Olivia Q. Antao
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Kindra M. Kelly‐Scumpia
- Division of CardiologyDepartment of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Sevana Baghdasarian
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Daniel P. Mayer
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Lily Shang
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Gina M. Sanchez
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Maani M. Archang
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
- MSTP ProgramDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Philip O. Scumpia
- Division of DermatologyDepartment of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of DermatologyVA Greater Los Angeles Healthcare SystemLos AngelesCA90073USA
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jason S Weinstein
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Dino Di Carlo
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Mechanical and Aerospace EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
- California Nano Systems Institute (CNSI)University of California Los AngelesLos AngelesCA90095USA
| |
Collapse
|
6
|
Xiao C, Li G, Li X, Wang D, Wu Y, Sun M, Huang J, Si L. A topical thermosensitive hydrogel system with cyclosporine A PEG-PCL micelles alleviates ulcerative colitis induced by TNBS in mice. Drug Deliv Transl Res 2023; 13:2447-2462. [PMID: 37060530 DOI: 10.1007/s13346-023-01317-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 04/16/2023]
Abstract
Ulcerative colitis (UC) is an idiopathic, chronic, relapsing disease. In most cases, only the distal colon is affected, and the colonic stasis or fast colonic transit through the inflamed colon usually results in reduced exposure of the distal inflamed colon. Although the immunosuppressant cyclosporine A (CsA) has been used in patients with severe colitis who do not respond to corticosteroids, the clinical application of CsA remains limited due to the systemic toxicities and insufficient accumulation at the site of action for the intravenous and oral routes. In this study, we loaded CsA into the amphipathic poly(ethylene glycol)-poly(ε-caprolactone) (PEG-PCL) micelles and then embedded them in hydrogels consisting of chitosan, poloxamer 188, and poloxamer 407 to construct a thermosensitive and mucoadhesive hydrogel drug delivery system (PLCP). The PLCP presented a high drug-loading capacity and showed a stable and rapid gelation rate after rectal administration into the body. Compared to CsA-loaded micelles and Sandimmun (Neoral®), the developed thermosensitive gel exhibited prolonged retention on the inflamed colon, as seen from in vitro adhesion and in vivo distribution experiments. It also fast mitigated colitis symptoms in TNBS-treated mice by regulating the expression levels of proinflammatory cytokines (TNF-α, IL-1β, COX-2, and iNOS2), anti-inflammatory cytokines (IL-10, Nrf2, NQO1, and HO-1), and other relevant biochemical factors. Our results suggested that CsA-loaded micelle thermal hydrogel system could be a promising strategy by enhancing the retention in the diseased colon and promoting the relief and recovery of UC.
Collapse
Affiliation(s)
- Chuyao Xiao
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Genyun Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoyue Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Minghui Sun
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiangeng Huang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luqin Si
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
7
|
Luo J, Zhao X, Guo B, Han Y. Preparation, thermal response mechanisms and biomedical applications of thermosensitive hydrogels for drug delivery. Expert Opin Drug Deliv 2023; 20:641-672. [PMID: 37218585 DOI: 10.1080/17425247.2023.2217377] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
INTRODUCTION Drug treatment is one of the main ways of coping with disease today. For the disadvantages of drug management, thermosensitive hydrogel is used as a countermeasure, which can realize the simple sustained release of drugs and the controlled release of drugs in complex physiological environments. AREAS COVERED This paper talks about thermosensitive hydrogels that can be used as drug carriers. The common preparation materials, material forms, thermal response mechanisms, characteristics of thermosensitive hydrogels for drug release and main disease treatment applications are reviewed. EXPERT OPINION When thermosensitive hydrogels are used as drug loading and delivery platforms, desired drug release patterns and release profiles can be tailored by selecting raw materials, thermal response mechanisms, and material forms. The properties of hydrogels prepared from synthetic polymers will be more stable than natural polymers. Integrating multiple thermosensitive mechanisms or different kinds of thermosensitive mechanisms on the same hydrogel is expected to realize the spatiotemporal differential delivery of multiple drugs under temperature stimulation. The industrial transformation of thermosensitive hydrogels as drug delivery platforms needs to meet some important conditions.
Collapse
Affiliation(s)
- Jinlong Luo
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xin Zhao
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Baolin Guo
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yong Han
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Affonso de Oliveira JF, Zhao Z, Xiang Y, Shin MD, Villaseñor KE, Deng X, Shukla S, Chen S, Steinmetz NF. COVID-19 vaccines based on viral nanoparticles displaying a conserved B-cell epitope show potent immunogenicity and a long-lasting antibody response. Front Microbiol 2023; 14:1117494. [PMID: 37152732 PMCID: PMC10157238 DOI: 10.3389/fmicb.2023.1117494] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/13/2023] [Indexed: 05/09/2023] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 sparked intensive research into the development of effective vaccines, 50 of which have been approved thus far, including the novel mRNA-based vaccines developed by Pfizer and Moderna. Although limiting the severity of the disease, the mRNA-based vaccines presented drawbacks, such as the cold chain requirement. Moreover, antibody levels generated by these vaccines decline significantly after 6 months. These vaccines deliver mRNA encoding the full-length spike (S) glycoprotein of SARS-CoV-2, but must be updated as new strains and variants of concern emerge, creating a demand for adjusted formulations and booster campaigns. To overcome these challenges, we have developed COVID-19 vaccine candidates based on the highly conserved SARS CoV-2, 809-826 B-cell peptide epitope (denoted 826) conjugated to cowpea mosaic virus (CPMV) nanoparticles and bacteriophage Qβ virus-like particles, both platforms have exceptional thermal stability and facilitate epitope delivery with inbuilt adjuvant activity. We evaluated two administration methods: subcutaneous injection and an implantable polymeric scaffold. Mice received a prime-boost regimen of 100 μg per dose (2 weeks apart) or a single dose of 200 μg administered as a liquid formulation, or a polymer implant. Antibody titers were evaluated longitudinally over 50 weeks. The vaccine candidates generally elicited an early Th2-biased immune response, which stimulates the production of SARS-CoV-2 neutralizing antibodies, followed by a switch to a Th1-biased response for most formulations. Exceptionally, vaccine candidate 826-CPMV (administered as prime-boost, soluble injection) elicited a balanced Th1/Th2 immune response, which is necessary to prevent pulmonary immunopathology associated with Th2 bias extremes. While the Qβ-based vaccine elicited overall higher antibody titers, the CPMV-induced antibodies had higher avidity. Regardless of the administration route and formulation, our vaccine candidates maintained high antibody titers for more than 50 weeks, confirming a potent and durable immune response against SARS-CoV-2 even after a single dose.
Collapse
Affiliation(s)
| | - Zhongchao Zhao
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | - Yi Xiang
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Matthew D. Shin
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | | | - Xinyi Deng
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Sourabh Shukla
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
| | - Shaochen Chen
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA, United States
- Center for Engineering in Cancer, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
9
|
Mahmudi H, Adili-Aghdam MA, Shahpouri M, Jaymand M, Amoozgar Z, Jahanban-Esfahlan R. Tumor microenvironment penetrating chitosan nanoparticles for elimination of cancer relapse and minimal residual disease. Front Oncol 2022; 12:1054029. [PMID: 36531004 PMCID: PMC9751059 DOI: 10.3389/fonc.2022.1054029] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/09/2022] [Indexed: 10/17/2023] Open
Abstract
Chitosan and its derivatives are among biomaterials with numerous medical applications, especially in cancer. Chitosan is amenable to forming innumerable shapes such as micelles, niosomes, hydrogels, nanoparticles, and scaffolds, among others. Chitosan derivatives can also bring unprecedented potential to cross numerous biological barriers. Combined with other biomaterials, hybrid and multitasking chitosan-based systems can be realized for many applications. These include controlled drug release, targeted drug delivery, post-surgery implants (immunovaccines), theranostics, biosensing of tumor-derived circulating materials, multimodal systems, and combination therapy platforms with the potential to eliminate bulk tumors as well as lingering tumor cells to treat minimal residual disease (MRD) and recurrent cancer. We first introduce different formats, derivatives, and properties of chitosan. Next, given the barriers to therapeutic efficacy in solid tumors, we review advanced formulations of chitosan modules as efficient drug delivery systems to overcome tumor heterogeneity, multi-drug resistance, MRD, and metastasis. Finally, we discuss chitosan NPs for clinical translation and treatment of recurrent cancer and their future perspective.
Collapse
Affiliation(s)
- Hossein Mahmudi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amin Adili-Aghdam
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Shahpouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Deliogullari B, Ilhan‐Ayisigi E, Cakmak B, Saglam‐Metiner P, Kaya N, Coskun‐Akar G, Yesil‐Celiktas O. Synthesis of an injectable heparin conjugated poloxamer hydrogel with high elastic recoverability for temporomandibular joint disorders. J Appl Polym Sci 2022. [DOI: 10.1002/app.52736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Buse Deliogullari
- Biomedical Technologies Graduate Programme, Graduate School of Natural and Applied Sciences Ege University Bornova Izmir Turkey
| | - Esra Ilhan‐Ayisigi
- Department of Bioengineering, Faculty of Engineering Ege University Izmir Turkey
- Genetic and Bioengineering Department, Faculty of Engineering and Architecture Kirsehir Ahi Evran University Kirsehir Turkey
| | - Betul Cakmak
- Department of Bioengineering, Faculty of Engineering Ege University Izmir Turkey
| | - Pelin Saglam‐Metiner
- Department of Bioengineering, Faculty of Engineering Ege University Izmir Turkey
| | - Nusret Kaya
- Department of Materials Science and Engineering, Faculty of Engineering and Architecture Izmir Katip Celebi University Cigli Izmir Turkey
| | - Gulcan Coskun‐Akar
- Department of Prosthodontics, Faculty of Dentistry Ege University Izmir Turkey
| | - Ozlem Yesil‐Celiktas
- Biomedical Technologies Graduate Programme, Graduate School of Natural and Applied Sciences Ege University Bornova Izmir Turkey
- Department of Bioengineering, Faculty of Engineering Ege University Izmir Turkey
| |
Collapse
|
11
|
Machado AS, Lage DP, Vale DL, Freitas CS, Linhares FP, Cardoso JM, Pereira IA, Ramos FF, Tavares GS, Ludolf F, Oliveira-da-Silva JA, Bandeira RS, Simões AC, Duarte MC, Oliveira JS, Christodoulides M, Chávez-Fumagalli MA, Roatt BM, Martins VT, Coelho EA. A recombinant Leishmania amastigote-specific protein, rLiHyG, with adjuvants, protects against infection with Leishmania infantum. Acta Trop 2022; 230:106412. [PMID: 35305943 DOI: 10.1016/j.actatropica.2022.106412] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022]
Abstract
Vaccination against visceral leishmaniasis (VL) should be considered as a control measure to protect against disease, and amastigote-specific proteins could help to develop such vaccines, since this parasite form is in contact with the host immune system during the active disease. In this study, a Leishmania amastigote-specific protein, LiHyG, was evaluated as recombinant protein (rLiHyG) as vaccine candidate against Leishmania infantum infection in BALB/c mice. The protein was associated with saponin (rLiHyG/Sap) or Poloxamer 407-based polymeric micelles (rLiHyG/Mic) as adjuvants, and animals receiving saline, saponin or micelle as controls. Immunological and parasitological analyses were performed before (n = 8 per group; as primary endpoint) and after (n = 8 per group; as secondary endpoint) infection. Results showed that, in both endpoints, rLiHyG/Sap and rLiHyG/Mic induced higher levels of IFN-γ, IL-12 and GM-CSF in spleen cell cultures from vaccinated animals, besides elevated presence of IgG2a isotype antibodies. Decreased hepatotoxicity and 'positive lymphoproliferative response were also found after challenge. Such findings reflected in significantly lower levels of parasite load found in their spleens, livers, bone marrows and draining lymph nodes. In conclusion, rLiHyG associated with Th1-type adjuvant could be considered for future studies as vaccine candidate to protect against VL.
Collapse
|
12
|
Lage DP, Machado AS, Vale DL, Freitas CS, Linhares FP, Cardoso JMO, Pereira IAG, Ramos FF, Tavares GSV, Ludolf F, Oliveira-da-Silva JA, Bandeira RS, Silva AM, Simões LC, Reis TAR, Oliveira JS, Christodoulides M, Chávez-Fumagalli MA, Roatt BM, Martins VT, Coelho EAF. Recombinant guanosine-5'-triphosphate (GTP)-binding protein associated with Poloxamer 407-based polymeric micelles protects against Leishmania infantum infection. Cytokine 2022; 153:155865. [PMID: 35339043 DOI: 10.1016/j.cyto.2022.155865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/12/2022] [Accepted: 03/09/2022] [Indexed: 11/03/2022]
Abstract
Leishmania virulence proteins should be considered as vaccine candidates against disease, since they are involved in developing infection in mammalian hosts. In a previous study, a Leishmania guanosine-5'-triphosphate (GTP)-binding protein was identified as a potential parasite virulence factor. In the present work, the gene encoding GTP was cloned and the recombinant protein (rGTP) was evaluated as a vaccine candidate against Leishmania infantum infection. The protein was associated with saponin (rGTP/Sap) or Poloxamer 407-based micelles (rGTP/Mic) as adjuvants, and protective efficacy was investigated in BALB/c mice after parasite challenge. Both rGTP/Sap and rGTP/Mic compositions induced a Th1-type immune response in vaccinated animals, with significantly higher levels of IFN-γ, IL-12, IL-2, TNF-α, GM-CSF, nitrite, specific IgG2a isotype antibody and positive lymphoproliferation, when compared to the control groups. This response was accompanied by significantly lower parasite load in the spleens, livers, bone marrows and draining lymph nodes of the animals. Immunological and parasitological evaluations indicated that rGTP/Mic induced a more polarized Th1-type response and higher reduction in the organ parasitism, and with lower hepatotoxicity, when compared to the use of rGTP/Sap. In conclusion, our preliminary data suggest that rGTP could be considered for further development as a vaccine candidate to protect against VL.
Collapse
Affiliation(s)
- Daniela P Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Amanda S Machado
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Danniele L Vale
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Camila S Freitas
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Flávia P Linhares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Jamille M O Cardoso
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Isabela A G Pereira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda F Ramos
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Ludolf
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - João A Oliveira-da-Silva
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Raquel S Bandeira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Alessandra M Silva
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana C Simões
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Thiago A R Reis
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Jamil S Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Myron Christodoulides
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton SO16 6YD, England
| | | | - Bruno M Roatt
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Vívian T Martins
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Prof. Alfredo Balena, 190, 30130-100, Belo Horizonte, Minas Gerais, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
13
|
Dmour I, Islam N. Recent advances on chitosan as an adjuvant for vaccine delivery. Int J Biol Macromol 2022; 200:498-519. [PMID: 34973993 DOI: 10.1016/j.ijbiomac.2021.12.129] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/05/2021] [Accepted: 12/19/2021] [Indexed: 12/21/2022]
Abstract
Chitosan (CS) is a natural polymer derived from chitin that has wide applications in drugs, vaccines, and antigen delivery. The distinctive mucoadhesive, biocompatibility, biodegradable, and less toxic properties of chitosan compared to the currently used vaccine adjuvants made it a promising candidate for use as an adjuvant/carrier in vaccine delivery. In addition, chitosan exhibits intrinsic immunomodulating properties making it a suitable adjuvant in preparing vaccines delivery systems. Nanoparticles (NPs) of chitosan and its derivatives loaded with antigen have been shown to induce cellular and humoral responses. Versatility in the physicochemical properties of chitosan can provide an excellent opportunity to engineer antigen-specific adjuvant/delivery systems. This review discusses the recent advances of chitosan and its derivatives as adjuvants in vaccine deliveryand the published literature in the last fifteen years. The impact of physicochemical properties of chitosan on vaccine formulation has been described in detail. Applications of chitosan and its derivatives, their physicochemical properties, and mechanisms in enhancing immune responses have been discussed. Finally, challenges and future aspects of chitosan use has been pointed out.
Collapse
Affiliation(s)
- Isra Dmour
- Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan.
| | - Nazrul Islam
- Pharmacy Discipline, School of Clinical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; Centre for Immunology and Infection Control (CIIC), Queensland University of Technology (QUT), Brisbane, QLD, Australia
| |
Collapse
|
14
|
Thermosensitive Poloxamer- graft-Carboxymethyl Pullulan: A Potential Injectable Hydrogel for Drug Delivery. Polymers (Basel) 2021; 13:polym13183025. [PMID: 34577926 PMCID: PMC8466796 DOI: 10.3390/polym13183025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/26/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
A thermosensitive copolymer composed of amphiphilic triblock copolymer, poloxamer 407, grafted on hydrophilic pullulan with pendant carboxymethyl groups (CMP) was prepared and characterized. The structure of the new copolymer was assessed by Fourier transform infrared (FT-IR) and 1H nuclear magnetic resonance (1H NMR) spectroscopy. The content of the poloxamer in the grafted copolymer was 83.8% (w/w). The effect of the copolymer concentration on the gelation behavior was analyzed by the vertical method and rheological tests; the gel phase of the copolymer occurred at a lower concentration (11%, w/v) as compared with poloxamer (18%, w/v). The starting gelation time under the simulated physiological conditions (phosphate buffer with a pH of 7.4, at 37 °C) was sensitive on the rest temperature before the test, this being 990 s and 280 s after 24 h rest at 4 °C and 20 °C, respectively. The rheological tests evidenced a high elasticity and excellent ability of the copolymer to recover the initial structure after the removal of the applied force or external stimuli. Moreover, the hydrogel has proved a sustained release of amoxicillin (taken as a model drug) over 168 h. Taken together, the results clearly indicate that this copolymer can be used as an injectable hydrogel.
Collapse
|
15
|
Luzuriaga MA, Shahrivarkevishahi A, Herbert FC, Wijesundara YH, Gassensmith JJ. Biomaterials and nanomaterials for sustained release vaccine delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1735. [PMID: 34180608 DOI: 10.1002/wnan.1735] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/03/2021] [Accepted: 06/04/2021] [Indexed: 12/17/2022]
Abstract
Vaccines are considered one of the most significant medical advancements in human history, as they have prevented hundreds of millions of deaths since their discovery; however, modern travel permits disease spread at unprecedented rates, and vaccine shortcomings like thermal sensitivity and required booster shots have been made evident by the COVID-19 pandemic. Approaches to overcoming these issues appear promising via the integration of vaccine technology with biomaterials, which offer sustained-release properties and preserve proteins, prevent conformational changes, and enable storage at room temperature. Sustained release and thermal stabilization of therapeutic biomacromolecules is an emerging area that integrates material science, chemistry, immunology, nanotechnology, and pathology to investigate different biocompatible materials. Biomaterials, including natural sugar polymers, synthetic polyesters produced from biologically derived monomers, hydrogel blends, protein-polymer blends, and metal-organic frameworks, have emerged as early players in the field. This overview will focus on significant advances of sustained release biomaterial in the context of vaccines against infectious disease and the progress made towards thermally stable "single-shot" formulations. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Michael A Luzuriaga
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Division of Medical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Fabian C Herbert
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardon, Texas, USA
| | - Yalini H Wijesundara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardon, Texas, USA
| | - Jeremiah J Gassensmith
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardon, Texas, USA.,Department of Bioengineering, The University of Texas at Dallas, Richardon, Texas, USA
| |
Collapse
|
16
|
Constantinou AP, Georgiou TK. Pre‐clinical and clinical applications of thermoreversible hydrogels in biomedical engineering: a review. POLYM INT 2021. [DOI: 10.1002/pi.6266] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Anna P Constantinou
- Department of Materials Imperial College London, South Kensington Campus, Royal School of Mines London UK
| | - Theoni K Georgiou
- Department of Materials Imperial College London, South Kensington Campus, Royal School of Mines London UK
| |
Collapse
|
17
|
Lima BV, Oliveira MJ, Barbosa MA, Gonçalves RM, Castro F. Immunomodulatory potential of chitosan-based materials for cancer therapy: a systematic review of in vitro, in vivo and clinical studies. Biomater Sci 2021; 9:3209-3227. [PMID: 33949372 DOI: 10.1039/d0bm01984d] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chitosan (Ch) has recently been used in different studies as a vaccine adjuvant with an ability to modulate the tumor microenvironment (TME). This systematic review aims to elucidate the added value of using Ch-based therapies for immunotherapeutic strategies in cancer treatment, through the exploration of different Ch-based formulations, their capacity to modulate immune cells in vitro and in vivo, and their translational potential for clinical settings. A systematic review was conducted on PubMed, following both inclusion and exclusion steps. Original articles which focused on the immunomodulatory role of Ch-based formulations in the TME were included, as well as its usage as a delivery vehicle for other immunomodulatory molecules. This review illustrates the added value of Ch-based systems to reshape the TME, through the modulation of immune cells using different Ch formulations, namely solutions, films, gels, microneedles and nanoparticles. Generally, Ch-based formulations increase the recruitment and proliferation of cells associated with pro-inflammatory abilities and decrease cells which exert anti-inflammatory activities. These effects correlated with a decreased tumor weight, reduced metastases, reversion of the immunosuppressive TME and increased survival in vivo. Overall, Ch-based formulations present the potential for immunotherapy in cancer. Nevertheless, clinical translation remains challenging, since the majority of the studies use Ch in formulations with other components, implicating that some of the observed effects could result from the combination of the individual effects. More studies on the use of different Ch-based formulations, complementary to standardization and disclosure of the Ch properties used are required to improve the immunomodulatory effects of Ch-based formulations in cancer.
Collapse
Affiliation(s)
- Beatriz V Lima
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal and ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Maria J Oliveira
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal and ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Mário A Barbosa
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal and ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Raquel M Gonçalves
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal and ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Flávia Castro
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal
| |
Collapse
|
18
|
Engineering a sustained release vaccine with a pathogen-mimicking manner for robust and durable immune responses. J Control Release 2021; 333:162-175. [PMID: 33794269 DOI: 10.1016/j.jconrel.2021.03.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Sustained release vaccine carriers can facilitate an increased interaction time between the antigen and immune system to strengthen immune responses, but their promotion on adaptive immune responses, especially cellular immunity, are still unfavorable. Herein, we report a sustained antigen delivery vector, which carries abundant antigens, a nucleic acid adjuvant and pathogen-associated molecular patterns to simulate a natural pathogen to reinforce immune responses. Specifically, murine colorectal cancer cells MC38 lysate and Toll-like receptor 9 agonist CpG are loaded into yeast derived β-glucan particles (GPs). After vaccination, these particles can form a vaccine depot that continuously release the antigen similar to the traditional aluminum hydroxide gel, but recruit more immune cells and induce more cytokine secretion at the injection site. Stronger antibody responses, Th1 and Th17 biased cellular immunity and immune memory are achieved compared with aluminum hydroxide gel. More importantly, treatment with these particles significantly suppress tumor growth in a therapeutic tumor model. This work shed light on the efficacy of combining sustained antigen release with pathogen-mimicking manner in vaccine design.
Collapse
|
19
|
Soni D, Bobbala S, Li S, Scott EA, Dowling DJ. The sixth revolution in pediatric vaccinology: immunoengineering and delivery systems. Pediatr Res 2021; 89:1364-1372. [PMID: 32927471 PMCID: PMC7511675 DOI: 10.1038/s41390-020-01112-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 05/08/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
Abstract
Infection is the predominant cause of mortality in early life, and immunization is the most promising biomedical intervention to reduce this burden. However, very young infants fail to respond optimally to most vaccines currently in use, especially neonates. In 2005, Stanley Plotkin proposed that new delivery systems would spur a new revolution in pediatric vaccinology, just as attenuation, inactivation, cell culture of viruses, genetic engineering, and adjuvantation had done in preceding decades. Recent advances in the field of immunoengineering, which is evolving alongside vaccinology, have begun to increasingly influence vaccine formulation design. Historically, the particulate nature of materials used in many vaccine formulations was empiric, often because of the need to stabilize antigens or reduce endotoxin levels. However, present vaccine delivery systems are rationally engineered to mimic the size, shape, and surface chemistry of pathogens, and are therefore often referred to as "pathogen-like particles". More than a decade from his original assessment, we re-assess Plotkin's prediction. In addition, we highlight how immunoengineering and advanced delivery systems may be uniquely capable of enhancing vaccine responses in vulnerable populations, such as infants. IMPACT: Immunoengineering and advanced delivery systems are leading to new developments in pediatric vaccinology. Summarizes delivery systems currently in use and development, and prospects for the future. Broad overview of immunoengineering's impact on vaccinology, catering to Pediatric Clinicians and Immunologists.
Collapse
Affiliation(s)
- Dheeraj Soni
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| | - Sharan Bobbala
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - Sophia Li
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - Evan A. Scott
- grid.16753.360000 0001 2299 3507Department of Biomedical Engineering, Northwestern University, Evanston, IL USA
| | - David J. Dowling
- grid.2515.30000 0004 0378 8438Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Boston, MA USA
| |
Collapse
|
20
|
Constantinou AP, Patias G, Somuncuoğlu B, Brock T, Lester DW, Haddleton DM, Georgiou TK. Homo- and co-polymerisation of di(propylene glycol) methyl ether methacrylate – a new monomer. Polym Chem 2021. [DOI: 10.1039/d1py00444a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A new methacrylate monomer with two propylene glycol groups on the side chain, di(propylene glycol) methyl ether methacrylate (diPGMA), was synthesised and homo- and co-polymerised for the first time.
Collapse
Affiliation(s)
| | | | | | - Toby Brock
- Department of Materials
- Imperial College London
- UK
| | | | | | | |
Collapse
|
21
|
Garcia-Del Rio L, Diaz-Rodriguez P, Landin M. Design of novel orotransmucosal vaccine-delivery platforms using artificial intelligence. Eur J Pharm Biopharm 2020; 159:36-43. [PMID: 33383169 DOI: 10.1016/j.ejpb.2020.12.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022]
Abstract
The linings of the oral cavity are excellent needle-free vaccination sites, able to induce immune responses at distal sites and confer systemic protection. However, owing to the mucosal tissues' intrinsic characteristics, the design of effective antigen-delivery systems is not an easy task. In the present work, we propose to develop and characterize thermosensitive and mucoadhesive hydrogels for orotransmucosal vaccination taking advantage of artificial intelligence tools (AIT). Hydrogels of variable composition were obtained combining Pluronic® F127 (PF127), Hybrane® S1200 (HS1200) and Gantrez® AN119 (AN119) or S97 (S97). Systems were characterized in terms of physicochemical properties, adhesion capacity to mucosal tissues and antigen-like microspheres release. Additionally, polymers biocompatibility and their immune-stimulation capacity was assessed in human macrophages. Interestingly, cells treated with HS1200 exhibited a significant proliferation enhancement compared to control. The use of AIT allowed to determine the effect of each polymer on formulations properties. The proportions of PF127 and Gantrez® are mainly the factors controlling gelation temperature, mucoadhesion, adhesion work and gel strength. Meanwhile, cohesion and short-term microsphere release are dependent on the PF127 concentration. However, long-term microsphere release varies depending on the Gantrez® variety and the PF127 concentration used. Hydrogels prepared with S97 showed slower microsphere release. The use of AIT allowed to establish the conditions able to produce ternary hydrogels with immune-stimulatory properties together with adequate mucoadhesion capacity and antigen-like microspheres release.
Collapse
Affiliation(s)
- Lorena Garcia-Del Rio
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), AeMat, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), IDIS Research Institute, 15706 Santiago de Compostela, Spain
| | - Patricia Diaz-Rodriguez
- Departamento de Ingeniería Química y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de La Laguna, Campus Anchieta, La Laguna 38200, Spain; Institute of Biomedical Technologies (ITB), Center for Biomedical Research of the Canary Islands (CIBICAN), Universidad de La Laguna, 38200 La Laguna, Spain.
| | - Mariana Landin
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), AeMat, Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), IDIS Research Institute, 15706 Santiago de Compostela, Spain
| |
Collapse
|
22
|
Vaccine implants: current status and recent advancements. Emerg Top Life Sci 2020; 4:319-330. [DOI: 10.1042/etls20200164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 01/29/2023]
Abstract
Implants have long been used in the field of drug delivery as controlled release vehicles and are now being investigated as single-shot vaccine technologies. Implants have shown great promise, minimizing the need for multiple immunizations while stimulating potent immune responses with reduced doses of vaccine. Synchronous release of vaccine components from implants over an appropriate period of time is important in order to avoid issues including immune tolerance, sequestration or deletion. Traditionally, implants require surgical implantation and removal, which can be a barrier to their widespread use. Degradable and in situ implants are now being developed that can be administered using minimally invasive subcutaneous or intramuscular injection techniques. Injectable hydrogels remain the most commonly studied approach for sustained vaccine delivery due to their ease of administration and tunable degradation properties. Despite exciting advancements in the field of vaccine implants, few technologies have progressed to clinical trials. To increase the likelihood of clinical translation of vaccine implants, strategic testing of disease-relevant antigens in appropriate species is essential. In this review, the significance of vaccine implants and the different types of implants being developed to deliver vaccines are discussed.
Collapse
|
23
|
Garcia-Vello P, Speciale I, Chiodo F, Molinaro A, De Castro C. Carbohydrate-based adjuvants. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 35-36:57-68. [PMID: 33388128 DOI: 10.1016/j.ddtec.2020.09.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/28/2020] [Accepted: 09/25/2020] [Indexed: 06/12/2023]
Abstract
Carbohydrate adjuvants are safe and biocompatible compounds usable as sustained delivery systems and stimulants of ongoing humoral and cellular immune responses, being especially suitable for the development of vaccines against intracellular pathogens where alum is useless. The development of new adjuvants is difficult and expensive, however, in the last two years, seven new carbohydrate-based adjuvants have been patented, also there are twelve ongoing clinical trials of vaccines that contain carbohydrate-based adjuvants, as well as numerous publications on their mechanism of action and safety. More research is necessary to improve the existent adjuvants and develop innovative ones.
Collapse
Affiliation(s)
- Pilar Garcia-Vello
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Naples (NA), Italy.
| | - Immacolata Speciale
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Naples (NA), Italy
| | - Fabrizio Chiodo
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, 80126 Naples (NA), Italy
| | - Cristina De Castro
- Department of Agricultural Sciences, University of Naples Federico II, Via Università, 100, 80055 Portici (NA), Italy.
| |
Collapse
|
24
|
Tricomponent thermoresponsive polymers based on an amine-containing monomer with tuneable hydrophobicity: Effect of composition. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Chen Z, Zhu Q, Qi J, Lu Y, Wu W. Sustained and controlled release of herbal medicines: The concept of synchronized release. Int J Pharm 2019; 560:116-125. [DOI: 10.1016/j.ijpharm.2019.01.074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 10/27/2022]
|
26
|
Allen SD, Bobbala S, Karabin NB, Scott EA. On the advancement of polymeric bicontinuous nanospheres toward biomedical applications. NANOSCALE HORIZONS 2019; 4:258-272. [PMID: 32254084 DOI: 10.1039/c8nh00300a] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Self-assembled soft nanocarriers that are capable of simultaneous encapsulation of both lipophilic and water soluble payloads have significantly enhanced controlled delivery applications in biomedicine. These nanoarchitectures, such as liposomes, polymersomes and cubosomes, are primarily composed of either amphiphilic polymers or lipids, with the polymeric variants generally possessing greater stability and control over biodistribution and bioresponsive release. Polymersomes have long demonstrated such advantages over their lipid analogs, liposomes, but only recently have bicontinuous nanospheres emerged as a polymeric cubic phase alternative to lipid cubosomes. In this review, we summarize the current state of the field for bicontinuous nanosphere formulation and characterization and suggest future directions for this nascent delivery platform as it is adopted for biomedical applications.
Collapse
Affiliation(s)
- Sean D Allen
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, USA.
| | | | | | | |
Collapse
|