1
|
Wang L, Ruan M, Bu Q, Zhao C. Signaling Pathways Driving MSC Osteogenesis: Mechanisms, Regulation, and Translational Applications. Int J Mol Sci 2025; 26:1311. [PMID: 39941080 PMCID: PMC11818554 DOI: 10.3390/ijms26031311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are crucial for skeletal development, homeostasis, and repair, primarily through their differentiation into osteoblasts and other skeletal lineage cells. Key signaling pathways, including Wnt, TGF-β/BMP, PTH, Hedgehog, and IGF, act as critical regulators of MSC osteogenesis, playing pivotal roles in maintaining bone homeostasis and facilitating regeneration. These pathways interact in distinct ways at various stages of bone development, mineralization, and remodeling. This review provides an overview of the molecular mechanisms by which these pathways regulate MSC osteogenesis, their influence on bone tissue formation, and their implications in bone diseases and therapeutic strategies. Additionally, we explore the potential applications of these pathways in bone tissue engineering, with a particular focus on promoting the use of MSCs as seed cells for bone defect repair. Ultimately, this review aims to highlight potential avenues for advancing bone biology research, treating bone disorders, and enhancing regenerative medicine.
Collapse
Affiliation(s)
| | | | | | - Chengzhu Zhao
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
2
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
3
|
Hou Y, Chen M, Bian Y, Zheng X, Tong R, Sun X. Advanced subunit vaccine delivery technologies: From vaccine cascade obstacles to design strategies. Acta Pharm Sin B 2023; 13:3321-3338. [PMID: 37655334 PMCID: PMC10465871 DOI: 10.1016/j.apsb.2023.01.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/23/2022] [Accepted: 12/03/2022] [Indexed: 01/12/2023] Open
Abstract
Designing and manufacturing safe and effective vaccines is a crucial challenge for human health worldwide. Research on adjuvant-based subunit vaccines is increasingly being explored to meet clinical needs. Nevertheless, the adaptive immune responses of subunit vaccines are still unfavorable, which may partially be attributed to the immune cascade obstacles and unsatisfactory vaccine design. An extended understanding of the crosstalk between vaccine delivery strategies and immunological mechanisms could provide scientific insight to optimize antigen delivery and improve vaccination efficacy. In this review, we summarized the advanced subunit vaccine delivery technologies from the perspective of vaccine cascade obstacles after administration. The engineered subunit vaccines with lymph node and specific cell targeting ability, antigen cross-presentation, T cell activation properties, and tailorable antigen release patterns may achieve effective immune protection with high precision, efficiency, and stability. We hope this review can provide rational design principles and inspire the exploitation of future subunit vaccines.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Min Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xi Zheng
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Kaur M, Nagpal M, Aggarwal G. Nanotechnology for Targeted Drug Delivery to Treat Osteoporosis. Curr Drug Targets 2023; 24:2-12. [PMID: 36200208 DOI: 10.2174/1389450123666221004124040] [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: 02/01/2022] [Revised: 05/13/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022]
Abstract
Bone diseases such as rheumatoid arthritis, Paget's disease, and osteoporosis cause mortality and mobility limits. Nanomedicine and nano delivery systems have been utilised to deliver active drug moiety to the precisely targeted site in a controlled manner, and it serves as a means of diagnostic tools. The utilisation of nanomedicine is expanding vigorously for assured targeting and efficient drug delivery. Nanotechnology offers various advantages, such as site-specific targeting, precise drug release kinetics, and improved bone mineral density. Recent medications available for osteoporosis are not viable due to the adverse effects associated with them and low patient compliance. There is an urgent need to develop biocompatible and appropriate drug delivery nanocarriers such as nanoparticles, liposomes, hydrogels, dendrimers, micelles, mesoporous particles, etc. These carriers enhance drug delivery and therapeutic effectiveness in bone tissues. The use of nanotechnology is also associated with toxicity. This article presents the review of various reports on nanocarrier systems and biologics for the treatment of osteoporosis. It aims to provide researchers with a clue for inventing a new drug delivery system with site-specific targeting for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Malkiet Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Geeta Aggarwal
- Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| |
Collapse
|
5
|
Nayak D, Chopra H, Chakrabartty I, Saravanan M, Barabadi H, Mohanta YK. Opportunities and challenges for bioengineered metallic nanoparticles as future nanomedicine. BIOENGINEERED NANOMATERIALS FOR WOUND HEALING AND INFECTION CONTROL 2023:517-540. [DOI: 10.1016/b978-0-323-95376-4.00012-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
6
|
Abstract
Polyanhydrides (PAs) are a class of synthetic biodegradable polymers employed as controlled drug delivery vehicles. They can be synthesized and scaled up from low-cost starting materials. The structure of PAs can be manipulated synthetically to meet desirable characteristics. PAs are biocompatible, biodegradable, and generate nontoxic metabolites upon degradation, which are easily eliminated from the body. The rate of water penetrating into the polyanhydride (PA) matrix is slower than the anhydride bond cleavage. This phenomenon sets PAs as "surface-eroding drug delivery carriers." Consequently, a variety of PA-based drug delivery carriers in the form of solid implants, pasty injectable formulations, microspheres, nanoparticles, etc. have been developed for the sustained release of small molecule drugs, and vaccines, peptide drugs, and nucleic acid-based active agents. The rate of drug delivery is often controlled by the polymer erosion rate, which is influenced by the polymer structure and composition, crystallinity, hydrophobicity, pH of the release medium, device size, configuration, etc. Owing to the above-mentioned interesting physicochemical and mechanical properties of PAs, the present review focuses on the advancements made in the domain of synthetic biodegradable biomedical PAs for therapeutic delivery applications. Various classes of PAs, their structures, their unique characteristics, their physicochemical and mechanical properties, and factors influencing surface erosion are discussed in detail. The review also summarizes various methods involved in the synthesis of PAs and their utility in the biomedical domain as drug, vaccine, and peptide delivery carriers in different formulations are reviewed.
Collapse
Affiliation(s)
- Pulikanti Guruprasad Reddy
- School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, and Centre for Cannabis Research and the Institute of Drug Research, The Alex Grass Centre for Drug Design and Synthesis, Jerusalem 9112002, Israel
| | - Abraham J Domb
- School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, and Centre for Cannabis Research and the Institute of Drug Research, The Alex Grass Centre for Drug Design and Synthesis, Jerusalem 9112002, Israel
| |
Collapse
|
7
|
Sarangi MK, Padhi S, Rath G, Nanda SS, Yi DK. Success of nano-vaccines against COVID-19: a transformation in nanomedicine. Expert Rev Vaccines 2022; 21:1739-1761. [PMID: 36384360 DOI: 10.1080/14760584.2022.2148659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The vaccines being used against COVID-19 are composed of either non-viral or viral nanoparticles (NPs). Nanotechnology-based vaccine technology was studied for its potentially transformative advancement of medicine. AREAS COVERED NPs protect the encapsulated mRNA in vaccines, thereby enhancing the stability of the ribonucleic acids and facilitating their intact delivery to their specific targets. Compared to liposomes, lipid nanoparticles (LNPs) are unique and, through their rigid morphology and better cellular penetrability, render enhanced cargo stability. To explore nanotechnology-mediated vaccine delivery and its potential in future pandemics, we assessed articles from various databases, such as PubMed, Embase, and Scopus, including editorial/research notes, expert opinions, and collections of data from several clinical research trials. In the current review, we focus on the nanoparticulate approach of the different SARS-CoV-2 vaccines and explore their success against the pandemic. EXPERT OPINION The mRNA-based vaccines, with their tremendous efficacy of ~95% (under phase III-IV clinical trials) and distinct nanocarriers (LNPs), represent a new medical front alongside DNA and siRNA-based vaccines.
Collapse
Affiliation(s)
- Manoj Kumar Sarangi
- Department of Pharmacy, School of Pharmaceutical Sciences, Sardar Bhagwan Singh University, Dehradun, India
| | - Sasmita Padhi
- Department of Pharmacy, School of Pharmaceutical Sciences, Sardar Bhagwan Singh University, Dehradun, India
| | - Gautam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, India
| | | | - Dong Kee Yi
- Department of Chemistry, Myongji University, Yongin, South Korea
| |
Collapse
|
8
|
Highly Stable Gold Nanoparticle-Antigen Conjugates with Self-Adjuvanting Property for Induction of Robust Antigen-Specific Immune Responses. Colloids Surf B Biointerfaces 2022; 220:112897. [DOI: 10.1016/j.colsurfb.2022.112897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/29/2022] [Accepted: 10/01/2022] [Indexed: 11/30/2022]
|
9
|
Sa-nguanmoo N, Namdee K, Khongkow M, Ruktanonchai U, Zhao Y, Liang XJ. Review: Development of SARS-CoV-2 immuno-enhanced COVID-19 vaccines with nano-platform. NANO RESEARCH 2021; 15:2196-2225. [PMID: 34659650 PMCID: PMC8501370 DOI: 10.1007/s12274-021-3832-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 05/04/2023]
Abstract
Vaccination is the most effective way to prevent coronavirus disease 2019 (COVID-19). Vaccine development approaches consist of viral vector vaccines, DNA vaccine, RNA vaccine, live attenuated virus, and recombinant proteins, which elicit a specific immune response. The use of nanoparticles displaying antigen is one of the alternative approaches to conventional vaccines. This is due to the fact that nano-based vaccines are stable, able to target, form images, and offer an opportunity to enhance the immune responses. The diameters of ultrafine nanoparticles are in the range of 1-100 nm. The application of nanotechnology on vaccine design provides precise fabrication of nanomaterials with desirable properties and ability to eliminate undesirable features. To be successful, nanomaterials must be uptaken into the cell, especially into the target and able to modulate cellular functions at the subcellular levels. The advantages of nano-based vaccines are the ability to protect a cargo such as RNA, DNA, protein, or synthesis substance and have enhanced stability in a broad range of pH, ambient temperatures, and humidity for long-term storage. Moreover, nano-based vaccines can be engineered to overcome biological barriers such as nonspecific distribution in order to elicit functions in antigen presenting cells. In this review, we will summarize on the developing COVID-19 vaccine strategies and how the nanotechnology can enhance antigen presentation and strong immunogenicity using advanced technology in nanocarrier to deliver antigens. The discussion about their safe, effective, and affordable vaccines to immunize against COVID-19 will be highlighted.
Collapse
Affiliation(s)
- Nawamin Sa-nguanmoo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Uracha Ruktanonchai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - YongXiang Zhao
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumour Theranostics and Therapy, Guangxi Medical University, Nanning, 530021 China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
10
|
Liu L, Kshirsagar P, Christiansen J, Gautam SK, Aithal A, Gulati M, Kumar S, Solheim JC, Batra SK, Jain M, Wannemuehler MJ, Narasimhan B. Polyanhydride nanoparticles stabilize pancreatic cancer antigen MUC4β. J Biomed Mater Res A 2021; 109:893-902. [PMID: 32776461 PMCID: PMC8100985 DOI: 10.1002/jbm.a.37080] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer (PC) is one of the most lethal malignancies and represents an increasing and challenging threat, especially with an aging population. The identification of immunogenic PC-specific upregulated antigens and an enhanced understanding of the immunosuppressive tumor microenvironment have provided opportunities to enable the immune system to recognize cancer cells. Due to its differential upregulation and functional role in PC, the transmembrane mucin MUC4 is an attractive target for immunotherapy. In the current study we characterized the antigen stability, antigenicity and release kinetics of a MUC4β-nanovaccine to guide further optimization and, in vivo evaluation. Amphiphilic polyanhydride copolymers based on 20 mol % 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane and 80 mol % 1,6-bis(p-carboxyphenoxy)hexane were used to synthesize nanoparticles. Structurally stable MUC4β protein was released from the particles in a sustained manner and characterized by gel electrophoresis and fluorescence spectroscopy. Modest levels of protein degradation were observed upon release. The released protein was also analyzed by MUC4β-specific monoclonal antibodies using ELISA and showed no significant loss of epitope availability. Further, mice immunized with multiple formulations of combination vaccines containing MUC4β-loaded nanoparticles generated MUC4β-specific antibody responses. These results indicate that polyanhydride nanoparticles are viable MUC4β vaccine carriers, laying the foundation for evaluation of this platform for PC immunotherapy.
Collapse
Affiliation(s)
- Luman Liu
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa
| | - Prakash Kshirsagar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - John Christiansen
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, Iowa
| | - Shailendra K. Gautam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mansi Gulati
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Joyce C. Solheim
- Nanovaccine Institute, Iowa State University, Ames, Iowa
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Nanovaccine Institute, Iowa State University, Ames, Iowa
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Nanovaccine Institute, Iowa State University, Ames, Iowa
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michael J. Wannemuehler
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, Iowa
- Nanovaccine Institute, Iowa State University, Ames, Iowa
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa
- Nanovaccine Institute, Iowa State University, Ames, Iowa
| |
Collapse
|
11
|
Xie X, Hu Y, Ye T, Chen Y, Zhou L, Li F, Xi X, Wang S, He Y, Gao X, Wei W, Ma G, Li Y. Therapeutic vaccination against leukaemia via the sustained release of co-encapsulated anti-PD-1 and a leukaemia-associated antigen. Nat Biomed Eng 2021; 5:414-428. [PMID: 33046865 DOI: 10.1038/s41551-020-00624-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/03/2020] [Indexed: 02/08/2023]
Abstract
Therapeutic leukaemia vaccines have shown modest potency. Here, we show that the co-encapsulation of a leukaemia-associated epitope peptide highly expressed in leukaemia patients and of the immune checkpoint inhibitor anti-programmed-cell-death-protein-1 (anti-PD-1) in degradable poly(lactic acid) microcapsules resulted in the sustained release of the peptide and of the antibody, which led to the recruitment of activated antigen-presenting cells to the injection site, their uptake of the peptide and the transportation of the anti-PD-1 antibody to lymph nodes, enhancing the expansion of epitope-specific T cells and the activation of cytotoxic T cells. After single subcutaneous injections of vaccine formulations with different epitope peptides, mice bearing leukaemia xenografts derived from humanized cell lines or from primary cells from patients showed better therapeutic outcomes than mice receiving repeated injections of free antigen, antibody and a commercial adjuvant. The sustained release of a tumour-associated peptide and of anti-PD-1 may represent a generalizable strategy for boosting antitumour immune responses to leukaemia.
Collapse
Affiliation(s)
- Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China.,State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Tong Ye
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Yiran Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Lijuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Feng Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Xiaobo Xi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Xiaoyong Gao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China. .,University of Chinese Academy of Sciences, Beijing, P R China.
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China. .,University of Chinese Academy of Sciences, Beijing, P R China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, P R China.
| |
Collapse
|
12
|
Mullis AS, Peroutka-Bigus N, Phadke KS, Bellaire BH, Narasimhan B. Nanomedicines to counter microbial barriers and antimicrobial resistance. Curr Opin Chem Eng 2021. [DOI: 10.1016/j.coche.2021.100672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Federizon J, Feugmo CGT, Huang WC, He X, Miura K, Razi A, Ortega J, Karttunen M, Lovell JF. Experimental and Computational Observations of Immunogenic Cobalt Porphyrin Lipid Bilayers: Nanodomain-Enhanced Antigen Association. Pharmaceutics 2021; 13:pharmaceutics13010098. [PMID: 33466686 PMCID: PMC7828809 DOI: 10.3390/pharmaceutics13010098] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/18/2022] Open
Abstract
Cobalt porphyrin phospholipid (CoPoP) can incorporate within bilayers to enable non-covalent surface-display of antigens on liposomes by mixing with proteins bearing a polyhistidine tag (his-tag); however, the mechanisms for how this occurs are poorly understood. These were investigated using the his-tagged model antigen Pfs25, a protein antigen candidate for malaria transmission-blocking vaccines. Pfs25 was found to associate with the small molecule aquocobalamin, a form of vitamin B12 and a cobalt-containing corrin macrocycle, but without particle formation, enabling comparative assessment. Relative to CoPoP liposomes, binding and serum stability studies indicated a weaker association of Pfs25 to aquocobalamin or cobalt nitrilotriacetic acid (Co-NTA) liposomes, which have cobalt displayed in the aqueous phase on lipid headgroups. Antigen internalization by macrophages was enhanced with Pfs25 bound to CoPoP liposomes. Immunization in mice with Pfs25 bound to CoPoP liposomes elicited antibodies that recognized ookinetes and showed transmission-reducing activity. To explore the physical mechanisms involved, we employed molecular dynamics (MD) simulations of bilayers containing phospholipid, cholesterol, as well as either CoPoP or NTA-functionalized lipids. The results show that the CoPoP-containing bilayer creates nanodomains that allow access for a limited but sufficient amount of water molecules that could be replaced by his-tags due to their favorable free energy properties allowing for stabilization. The position of the metal center within the NTA liposomes was much more exposed to the aqueous environment, which could explain its limited capacity for stabilizing Pfs25. This study illustrates the impact of CoPoP-induced antigen particleization in enhancing vaccine efficacy, and provides molecular insights into the CoPoP bilayer properties that enable this.
Collapse
Affiliation(s)
- Jasmin Federizon
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.F.); (W.-C.H.); (X.H.)
| | | | - Wei-Chiao Huang
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.F.); (W.-C.H.); (X.H.)
| | - Xuedan He
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.F.); (W.-C.H.); (X.H.)
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA;
| | - Aida Razi
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada; (A.R.); (J.O.)
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada; (A.R.); (J.O.)
| | - Mikko Karttunen
- Department of Chemistry, the University of Western Ontario, London, ON N6A 3K7, Canada;
- Centre for Advanced Materials and Biomaterials Research, the University of Western Ontario, London, ON N6A 3K7, Canada
- Department of Applied Mathematics, the University of Western Ontario, London, ON N6A 5B7, Canada
- Correspondence: (M.K.); (J.F.L.)
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA; (J.F.); (W.-C.H.); (X.H.)
- Correspondence: (M.K.); (J.F.L.)
| |
Collapse
|
14
|
Grego EA, Siddoway AC, Uz M, Liu L, Christiansen JC, Ross KA, Kelly SM, Mallapragada SK, Wannemuehler MJ, Narasimhan B. Polymeric Nanoparticle-Based Vaccine Adjuvants and Delivery Vehicles. Curr Top Microbiol Immunol 2021; 433:29-76. [PMID: 33165869 PMCID: PMC8107186 DOI: 10.1007/82_2020_226] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As vaccine formulations have progressed from including live or attenuated strains of pathogenic components for enhanced safety, developing new adjuvants to more effectively generate adaptive immune responses has become necessary. In this context, polymeric nanoparticles have emerged as a promising platform with multiple advantages, including the dual capability of adjuvant and delivery vehicle, administration via multiple routes, induction of rapid and long-lived immunity, greater shelf-life at elevated temperatures, and enhanced patient compliance. This comprehensive review describes advances in nanoparticle-based vaccines (i.e., nanovaccines) with a particular focus on polymeric particles as adjuvants and delivery vehicles. Examples of the nanovaccine approach in respiratory infections, biodefense, and cancer are discussed.
Collapse
Affiliation(s)
- Elizabeth A Grego
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Alaric C Siddoway
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Metin Uz
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Luman Liu
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - John C Christiansen
- Departments of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, 50011, USA
| | - Kathleen A Ross
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Sean M Kelly
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Surya K Mallapragada
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Michael J Wannemuehler
- Departments of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, 50011, USA
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA
| | - Balaji Narasimhan
- Departments of Chemical and Biological Engineering, Iowa State University, Ames, IA, 50011, USA.
- Departments of Nanovaccine Institute, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
15
|
Maina TW, Grego EA, Boggiatto PM, Sacco RE, Narasimhan B, McGill JL. Applications of Nanovaccines for Disease Prevention in Cattle. Front Bioeng Biotechnol 2020; 8:608050. [PMID: 33363134 PMCID: PMC7759628 DOI: 10.3389/fbioe.2020.608050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Vaccines are one of the most important tools available to prevent and reduce the incidence of infectious diseases in cattle. Despite their availability and widespread use to combat many important pathogens impacting cattle, several of these products demonstrate variable efficacy and safety in the field, require multiple doses, or are unstable under field conditions. Recently, nanoparticle-based vaccine platforms (nanovaccines) have emerged as promising alternatives to more traditional vaccine platforms. In particular, polymer-based nanovaccines provide sustained release of antigen payloads, stabilize such payloads, and induce enhanced antibod- and cell-mediated immune responses, both systemically and locally. To improve vaccine administrative strategies and efficacy, they can be formulated to contain multiple antigenic payloads and have the ability to protect fragile proteins from degradation. Nanovaccines are also stable at room temperature, minimizing the need for cold chain storage. Nanoparticle platforms can be synthesized for targeted delivery through intranasal, aerosol, or oral administration to induce desired mucosal immunity. In recent years, several nanovaccine platforms have emerged, based on biodegradable and biocompatible polymers, liposomes, and virus-like particles. While most nanovaccine candidates have not yet advanced beyond testing in rodent models, a growing number have shown promise for use against cattle infectious diseases. This review will highlight recent advancements in polymeric nanovaccine development and the mechanisms by which nanovaccines may interact with the bovine immune system. We will also discuss the positive implications of nanovaccines use for combating several important viral and bacterial disease syndromes and consider important future directions for nanovaccine development in beef and dairy cattle.
Collapse
Affiliation(s)
- Teresia W. Maina
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Elizabeth A. Grego
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Paola M. Boggiatto
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Randy E. Sacco
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
16
|
Biodistribution of degradable polyanhydride particles in Aedes aegypti tissues. PLoS Negl Trop Dis 2020; 14:e0008365. [PMID: 32898130 PMCID: PMC7500644 DOI: 10.1371/journal.pntd.0008365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/18/2020] [Accepted: 05/05/2020] [Indexed: 01/18/2023] Open
Abstract
Insecticide resistance poses a significant threat to the control of arthropods that transmit disease agents. Nanoparticle carriers offer exciting opportunities to expand the armamentarium of insecticides available for public health and other pests. Most chemical insecticides are delivered by contact or feeding, and from there must penetrate various biological membranes to reach target organs and kill the pest organism. Nanoparticles have been shown to improve bioactive compound navigation of such barriers in vertebrates, but have not been well-explored in arthropods. In this study, we explored the potential of polyanhydride micro- and nanoparticles (250 nm- 3 μm), labeled with rhodamine B to associate with and/or transit across insect biological barriers, including the cuticle, epithelium, midgut and ovaries, in female Ae. aeygpti mosquitoes. Mosquitoes were exposed using conditions to mimic surface contact with a residual spray or paint, topical exposure to mimic contact with aerosolized insecticide, or per os in a sugar meal. In surface contact experiments, microparticles were sometimes observed in association with the exterior of the insect cuticle. Nanoparticles were more uniformly distributed across exterior tissues and present at higher concentrations. Furthermore, by surface contact, topical exposure, or per os, particles were detected in internal organs. In every experiment, amphiphilic polyanhydride nanoparticles associated with internal tissues to a higher degree than hydrophobic nanoparticles. In vitro, nanoparticles associated with Aedes aegypti Aag2 cells within two hours of exposure, and particles were evident in the cytoplasm. Further studies demonstrated that particle uptake is dependent on caveolae-mediated endocytosis. The propensity of these nanoparticles to cross biological barriers including the cuticle, to localize in target tissue sites of interest, and to reach the cytoplasm of cells, provides great promise for targeted delivery of insecticidal candidates that cannot otherwise reach these cellular and subcellular locations.
Collapse
|
17
|
Mullis AS, Jacobson SJ, Narasimhan B. High-Throughput Synthesis and Screening of Rapidly Degrading Polyanhydride Nanoparticles. ACS COMBINATORIAL SCIENCE 2020; 22:172-183. [PMID: 32125826 DOI: 10.1021/acscombsci.9b00162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Combinatorial techniques can accelerate the discovery and development of polymeric nanodelivery devices by pairing high-throughput synthesis with rapid materials characterization. Biodegradable polyanhydrides demonstrate tunable release, high cellular internalization, and dose sparing properties when used as nanodelivery devices. This nanoparticle platform shows promising potential for small molecule drug delivery, but the pace of understanding and rational design of these nanomedicines is limited by the low throughput of conventional characterization. This study reports the use of a high-throughput method to synthesize libraries of a newly synthesized, rapidly eroding polyanhydride copolymer based on 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG) and sebacic acid (SA) monomers. The high-throughput method enabled efficient screening of copolymer microstructure, revealing weak block-type and alternating architectures. The high-throughput method was adapted to synthesize nanoparticle libraries encapsulating hydrophobic model drugs. Drug release from these nanoparticles was rapid, with a majority of the payload released within 3 days. Drug release was dramatically slowed at acidic pH, which could be useful for oral drug delivery. Rhodamine B (RhoB) release kinetics generally followed patterns of polymer erosion kinetics, while Coomassie brilliant blue (CBB) released the fastest from the slowest degrading polymer chemistry and vice versa. These differences in trends between copolymer chemistry and release kinetics were hypothesized to arise from differences in mixing thermodynamics. A high-throughput method was developed to synthesize polymer-drug film libraries and characterize mixing thermodynamics by melting point depression. Rhodamine B had a negative χ for all copolymers with <30 mol % CPTEG tested, indicating a tendency toward miscibility. By contrast, CBB χ increased, eventually becoming positive near 15:85 CPTEG:SA, with increasing CPTEG content. This indicates an increasing tendency toward phase separation in CPTEG-rich copolymers. These in vitro results screening polymer-drug interactions showed good agreement with in silico predictions from Hansen solubility parameter estimation and were able to explain the observed differences in model drug release trends.
Collapse
Affiliation(s)
- Adam S. Mullis
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Sarah J. Jacobson
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
18
|
Bakshi S, Sanz Garcia R, Van der Weken H, Tharad A, Pandey S, Juarez P, Virdi V, Devriendt B, Cox E, Depicker A. Evaluating single-domain antibodies as carriers for targeted vaccine delivery to the small intestinal epithelium. J Control Release 2020; 321:416-429. [PMID: 31981657 DOI: 10.1016/j.jconrel.2020.01.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/13/2020] [Accepted: 01/18/2020] [Indexed: 12/24/2022]
Abstract
Targeting a vaccine to the mucosal surface has recently been recognized as a promising approach to efficiently induce mucosal immune responses against enteric pathogens. However, poor uptake and inefficient transport of orally delivered subunit vaccines across the intestinal epithelium combined with weak immune responses still present important bottlenecks for mucosal vaccination. A possible strategy suggested to surmount these hurdles is to target the selected antigen to transcytotic receptors, such as aminopeptidase N (APN) present on enterocytes and antigen-presenting cells (APCs). Therefore, we aimed to identify potent and selective VHHs against porcine aminopeptidase N (pAPN), that were fused to the fragment crystallizable (Fc) domain of the murine IgG2a, resulting in dimeric VHH-MG fusions. Out of a library of 30 VHH-MG fusion candidates, two fusions displaying the best binding on pAPN-expressing cells were selected and showed in vivo internalization across the porcine gut epithelium. One of these fusions triggered systemic and intestinal IgA responses upon oral administration. Our results demonstrate the potential of bivalent VHH-MG fusions as delivery vehicles for vaccine antigens. VHH-mediated targeting of antigens to APN to generate protective immunity at the mucosal surface remains to be further validated.
Collapse
Affiliation(s)
- Shruti Bakshi
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Gent, Belgium; VIB Center for Plant Systems Biology, 9052 Gent, Belgium
| | - Raquel Sanz Garcia
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Hans Van der Weken
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Ashuwini Tharad
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Gent, Belgium; VIB Center for Plant Systems Biology, 9052 Gent, Belgium
| | - Shubham Pandey
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Gent, Belgium; VIB Center for Plant Systems Biology, 9052 Gent, Belgium
| | - Paloma Juarez
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Gent, Belgium; VIB Center for Plant Systems Biology, 9052 Gent, Belgium
| | - Vikram Virdi
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Gent, Belgium; VIB Center for Plant Systems Biology, 9052 Gent, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium.
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium.
| | - Ann Depicker
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Gent, Belgium; VIB Center for Plant Systems Biology, 9052 Gent, Belgium.
| |
Collapse
|
19
|
Kelly SM, Mitra A, Mathur S, Narasimhan B. Synthesis and Characterization of Rapidly Degrading Polyanhydrides as Vaccine Adjuvants. ACS Biomater Sci Eng 2020; 6:265-276. [PMID: 33463223 DOI: 10.1021/acsbiomaterials.9b01427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is a currently a need to develop adjuvants that are best suited to simultaneously enhance immune responses, induce immunologic memory, improve patient compliance (i.e., reduce doses and inflammation), and provide vaccine shelf stability for stockpiling and global deployment to challenging environments. Biodegradable polyanhydrides have been investigated extensively to overcome such challenges. It has been shown that controlling copolymer composition can result in chemistry-dependent immunomodulatory capabilities. These studies have revealed that copolymers rich in sebacic acid (SA) are highly internalized by antigen presenting cells and confer improved shelf stability of encapsulated proteins, while copolymers rich in 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG) also exhibit enhanced internalization by and activation of antigen presenting cells (APCs), in addition to providing superior retention of protein stability following encapsulation and release. However, to date, CPTEG:SA copolymers have not been synthesized and described. In this work, we hypothesized that new copolymers composed of CPTEG and SA would combine the advantages of both monomers in terms of enhanced thermal properties, maintaining antigenicity of encapsulated proteins following nanoparticle synthesis, and superior cellular internalization and activation by APCs, demonstrated by the upregulation of costimulatory markers CD80, CD86, and CD40, as well as the secretion of proinflammatory cytokines IL-6, IL-1β, and TNF-α. Herein, we describe the synthesis and design of novel CPTEG:SA nanoparticles with improved thermal properties, payload stability, and internalization by antigen presenting cells for applications in vaccine delivery. The performance of these new CPTEG:SA formulations was compared to that of traditional polyanhydride copolymers.
Collapse
Affiliation(s)
- Sean M Kelly
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Akash Mitra
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Srishti Mathur
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Iowa State University, Ames, Iowa 50011-1098, United States
| |
Collapse
|
20
|
Wagner DA, Kelly SM, Petersen AC, Peroutka-Bigus N, Darling RJ, Bellaire BH, Wannemuehler MJ, Narasimhan B. Single-dose combination nanovaccine induces both rapid and long-lived protection against pneumonic plague. Acta Biomater 2019; 100:326-337. [PMID: 31610342 PMCID: PMC7012387 DOI: 10.1016/j.actbio.2019.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 02/01/2023]
Abstract
Yersinia pestis, the causative agent of pneumonic plague, induces a highly lethal infection if left untreated. Currently, there is no FDA-approved vaccine against this pathogen; however, USAMRIID has developed a recombinant fusion protein, F1-V, that has been shown to induce protection against pneumonic plague. Many F1-V-based vaccine formulations require prime-boost immunization to achieve protective immunity, and there are limited reports of rapid induction of protective immunity (≤ 14 days post-immunization (DPI)). The STimulator of INterferon Genes agonists cyclic dinucleotides (CDNs) have been shown to be promising vaccine adjuvants. Polyanhydride nanoparticle-based vaccines (i.e., nanovaccines) have also shown to enhance immune responses due to their dual functionality as adjuvants and delivery vehicles. In this work, a combination nanovaccine was designed that comprised F1-V-loaded nanoparticles combined with the CDN, dithio-RP,RP-cyclic di-guanosine monophosphate, to induce rapid and long-lived protective immunity against pneumonic plague. All mice immunized with a single dose combination nanovaccine were protected from Y. pestis lethal challenge within 14 DPI and demonstrated enhanced protection over F1-V adjuvanted with CDNs alone at challenge doses ≥7000 CFU Y. pestis CO92. In addition, 75% of mice receiving the single dose of the combination nanovaccine were protected from challenge at 182 DPI, while maintaining high levels of antigen-specific serum IgG. ELISPOT analysis of vaccinated animals at 218 DPI revealed F1-V-specific long-lived plasma cells in bone marrow in mice vaccinated with CDN adjuvanted F1-V or the combination nanovaccine. Microarray analysis of serum from these vaccinated mice revealed the presence of serum antibody that bound to a broad range of F1 and V linear epitopes. These results demonstrate that combining the adjuvanticity of CDNs with a nanovaccine delivery system enables induction of both rapid and long-lived protective immunity against Y. pestis. STATEMENT OF SIGNIFICANCE: • Yersinia pestis, the causative agent of pneumonic plague, induces a highly lethal infection if left untreated. Currently, there is no FDA-approved vaccine against this biodefense pathogen. • We designed a combination nanovaccine comprising of F1-V antigen-loaded polyanhydride nanoparticles and a cyclic dinucleotide adjuvant to induce both rapid and long-lived protective immunity against pneumonic plague. • Animals immunized with the combination nanovaccine maintained high levels of antigen-specific serum IgG and long-lived plasma cells in bone marrow and the serum antibody showed a high affinity for a broad range of F1 and V linear epitopes. • The combination nanovaccine is a promising next-generation vaccine platform against weaponized Y. pestis based on its ability to induce both rapid and long-lived protective immunity.
Collapse
Affiliation(s)
- Danielle A Wagner
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Sean M Kelly
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Andrew C Petersen
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Nathan Peroutka-Bigus
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States; Interdepartmental Microbiology Program, Iowa State University, Ames, IA, United States
| | - Ross J Darling
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Bryan H Bellaire
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States; Interdepartmental Microbiology Program, Iowa State University, Ames, IA, United States; Nanovaccine Institute, Iowa State University, Ames, IA, United States
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States; Nanovaccine Institute, Iowa State University, Ames, IA, United States.
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States; Nanovaccine Institute, Iowa State University, Ames, IA, United States.
| |
Collapse
|
21
|
Madani F, Hsein H, Busignies V, Tchoreloff P. An overview on dosage forms and formulation strategies for vaccines and antibodies oral delivery. Pharm Dev Technol 2019; 25:133-148. [DOI: 10.1080/10837450.2019.1689402] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Yang G, Chen S, Zhang J. Bioinspired and Biomimetic Nanotherapies for the Treatment of Infectious Diseases. Front Pharmacol 2019; 10:751. [PMID: 31333467 PMCID: PMC6624236 DOI: 10.3389/fphar.2019.00751] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
There are still great challenges for the effective treatment of infectious diseases, although considerable achievement has been made by using antiviral and antimicrobial agents varying from small-molecule drugs, peptides/proteins, to nucleic acids. The nanomedicine approach is emerging as a new strategy capable of overcoming disadvantages of molecular therapeutics and amplifying their anti-infective activities, by localized delivery to infection sites, reducing off-target effects, and/or attenuating resistance development. Nanotechnology, in combination with bioinspired and biomimetic approaches, affords additional functions to nanoparticles derived from synthetic materials. Herein, we aim to provide a state-of-the-art review on recent progress in biomimetic and bioengineered nanotherapies for the treatment of infectious disease. Different biomimetic nanoparticles, derived from viruses, bacteria, and mammalian cells, are first described, with respect to their construction and biophysicochemical properties. Then, the applications of diverse biomimetic nanoparticles in anti-infective therapy are introduced, either by their intrinsic activity or by loading and site-specifically delivering various molecular drugs. Bioinspired and biomimetic nanovaccines for prevention and/or therapy of infectious diseases are also highlighted. At the end, major translation issues and future directions of this field are discussed.
Collapse
Affiliation(s)
- Guoyu Yang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
- The First Clinical College, Chongqing Medical University, Chongqing, China
| | - Sheng Chen
- Department of Pediatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
23
|
Klahn P, Fetz V, Ritter A, Collisi W, Hinkelmann B, Arnold T, Tegge W, Rox K, Hüttel S, Mohr KI, Wink J, Stadler M, Wissing J, Jänsch L, Brönstrup M. The nuclear export inhibitor aminoratjadone is a potent effector in extracellular-targeted drug conjugates. Chem Sci 2019; 10:5197-5210. [PMID: 31191875 PMCID: PMC6540907 DOI: 10.1039/c8sc05542d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/15/2019] [Indexed: 12/04/2022] Open
Abstract
The concept of targeted drug conjugates has been successfully translated to clinical practice in oncology. Whereas the majority of cytotoxic effectors in drug conjugates are directed against either DNA or tubulin, our study aimed to validate nuclear export inhibition as a novel effector principle in drug conjugates. For this purpose, a semisynthetic route starting from the natural product ratjadone A, a potent nuclear export inhibitor, has been developed. The biological evaluation of ratjadones functionalized at the 16-position revealed that oxo- and amino-analogues had very high potencies against cancer cell lines (e.g. 16R-aminoratjadone 16 with IC50 = 260 pM against MCF-7 cells, or 19-oxoratjadone 14 with IC50 = 100 pM against A-549 cells). Mechanistically, the conjugates retained a nuclear export inhibitory activity through binding CRM1. To demonstrate a proof-of-principle for cellular targeting, folate- and luteinizing hormone releasing hormone (LHRH)-based carrier molecules were synthesized and coupled to aminoratjadones as well as fluorescein for cellular efficacy and imaging studies, respectively. The Trojan-Horse conjugates selectively addressed receptor-positive cell lines and were highly potent inhibitors of their proliferation. For example, the folate conjugate FA-7-Val-Cit-pABA-16R-aminoratjadone had an IC50 of 34.3 nM, and the LHRH conjugate d-Orn-Gose-Val-Cit-pABA-16R-aminoratjadone had an IC50 of 12.8 nM. The results demonstrate that nuclear export inhibition is a promising mode-of-action for extracellular-targeted drug conjugate payloads.
Collapse
Affiliation(s)
- Philipp Klahn
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
- Institute of Organic Chemistry , Technische Universität Braunschweig , Hagenring 30 , 38106 Braunschweig , Germany .
| | - Verena Fetz
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
| | - Antje Ritter
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
| | - Wera Collisi
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
- Department of Microbial Drugs , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany
| | - Bettina Hinkelmann
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
| | - Tatjana Arnold
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
| | - Werner Tegge
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
| | - Katharina Rox
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
- German Centre of Infection Research (DZIF) , Partner Site Hannover-Braunschweig , Germany
| | - Stephan Hüttel
- Department of Microbial Drugs , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany
| | - Kathrin I Mohr
- Department of Microbial Drugs , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany
| | - Joachim Wink
- Department of Microbial Drugs , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany
| | - Marc Stadler
- Department of Microbial Drugs , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany
| | - Josef Wissing
- Department of Structure and Function of Proteins , Research Group Cellular Proteomic , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany
| | - Lothar Jänsch
- Department of Structure and Function of Proteins , Research Group Cellular Proteomic , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany
| | - Mark Brönstrup
- Department of Chemical Biology , Helmholtz Centre for Infection Research , Inhoffenstrasse 7 , 38124 Braunschweig , Germany .
- Biomolecular Drug Research Center (BMWZ) , Schneiderberg 38 , 30167 Hannover , Germany
- German Centre of Infection Research (DZIF) , Partner Site Hannover-Braunschweig , Germany
| |
Collapse
|
24
|
Adams JR, Senapati S, Haughney SL, Wannemuehler MJ, Narasimhan B, Mallapragada SK. Safety and biocompatibility of injectable vaccine adjuvants composed of thermogelling block copolymer gels. J Biomed Mater Res A 2019; 107:1754-1762. [PMID: 30972906 DOI: 10.1002/jbm.a.36691] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 02/25/2019] [Accepted: 03/22/2019] [Indexed: 11/08/2022]
Abstract
Injectable thermogelling polymers have been recently investigated as novel adjuvants and delivery systems for next generation vaccines. As research into natural and synthetic biocompatible polymers progresses, the safety and biocompatibility of these compounds is of paramount importance. We have developed cationic pentablock copolymer (PBC) vaccine adjuvants based on Pluronic F127, a thermogelling triblock copolymer that has been approved by the FDA for multiple applications, and methacrylated poly(diethyl amino)ethyl methacrylate outer blocks. These novel materials have been demonstrated to effectively create an antigen depot, minimally impact antigen stability, and enhance the immune response to antigens (i.e., adjuvanticity) in mice. In this work, we investigated the safety and biocompatibility of the parent triblock Pluronic gels and the cationic PBC gels in mice. Histological analysis showed no injection site reactions and no damage to the liver or kidneys was observed upon administering the block copolymer formulations. However, the subcutaneous injection of a thermogelling Pluronic solution induced increased levels of lipids in the blood, with no further deleterious effects observed from the addition of the cationic outer blocks. This hyperlipidemia resolved within 30 days after the administration of the Pluronic formulation. To mitigate this adverse effect, the vaccine adjuvant formulations were modified by adding poly(vinyl alcohol), which allowed gelation, while reducing the amount of Pluronic in the formulation. This modified formulation abrogated the observed hyperlipidemia and no adverse effects were observed in the serum through biomarker analysis or at the injection site (i.e., inflammation) in comparison to the responses induced by administration of saline or incomplete Freund's adjuvant. These studies provide a foundation to developing these gels as adjuvants for next generation vaccines. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1754-1762, 2019.
Collapse
Affiliation(s)
- Justin R Adams
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011
| | - Sujata Senapati
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011
| | - Shannon L Haughney
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa, 50011
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011
| |
Collapse
|
25
|
Mullis AS, Broderick SR, Binnebose AM, Peroutka-Bigus N, Bellaire BH, Rajan K, Narasimhan B. Data Analytics Approach for Rational Design of Nanomedicines with Programmable Drug Release. Mol Pharm 2019; 16:1917-1928. [PMID: 30973741 DOI: 10.1021/acs.molpharmaceut.8b01272] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Drug delivery vehicles can improve the functional efficacy of existing antimicrobial therapies by improving biodistribution and targeting. A critical property of such nanomedicine formulations is their ability to control the release kinetics of their payloads. The combination of (and interactions among) polymer, drug, and nanoparticle properties gives rise to nonlinear behavioral relationships and large data space. These factors complicate both first-principles modeling and screening of nanomedicine formulations. Predictive analytics may offer a more efficient approach toward the rational design of nanomedicines by identifying key descriptors and correlating them to nanoparticle release behavior. In this work, antibiotic release kinetics data were generated from polyanhydride nanoparticle formulations with varying copolymer compositions, encapsulated drug type, and drug loading. Four antibiotics, doxycycline, rifampicin, chloramphenicol, and pyrazinamide, were used. Linear manifold learning methods were used to relate drug release properties with polymer, drug, and nanoparticle properties, and key descriptors were identified that are highly correlated with release properties. However, these linear methods could not predict release behavior. Nonlinear multivariate modeling based on graph theory was then used to deconvolute the governing relationships between these properties, and predictive models were generated to rapidly screen lead nanomedicine formulations with desirable release properties with minimal nanoparticle characterization. Release kinetics predictions of two drugs containing atoms not included in the model showed good agreement with experimental results, validating the model and indicating its potential to virtually explore new polymer and drug pairs not included in the training data set. The models were shown to be robust after the inclusion of these new formulations, in that the new inclusions did not significantly change model regression. This approach provides the first step toward the development of a framework that can be used to rationally design nanomedicine formulations by selecting the appropriate carrier for a drug payload to program desirable release kinetics.
Collapse
Affiliation(s)
| | - Scott R Broderick
- Department of Materials Design and Innovation , University at Buffalo , Buffalo , New York 14260 , United States
| | | | | | | | - Krishna Rajan
- Department of Materials Design and Innovation , University at Buffalo , Buffalo , New York 14260 , United States
| | | |
Collapse
|
26
|
Senapati S, Darling RJ, Loh D, Schneider IC, Wannemuehler MJ, Narasimhan B, Mallapragada SK. Pentablock Copolymer Micelle Nanoadjuvants Enhance Cytosolic Delivery of Antigen and Improve Vaccine Efficacy while Inducing Low Inflammation. ACS Biomater Sci Eng 2019; 5:1332-1342. [PMID: 33405651 PMCID: PMC8627116 DOI: 10.1021/acsbiomaterials.8b01591] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
As the focus has shifted from traditional killed or live, attenuated vaccines toward subunit vaccines, improvements in vaccine safety have been confronted with low immunogenicity of protein antigens. This issue has been addressed by synthesizing and designing a wide variety of antigen carriers and adjuvants, such as Toll-like receptor agonists (e.g., MPLA, CpG). Studies have focused on optimizing adjuvants for improved cellular trafficking, cytosolic availability, and improved antigen presentation. In this work, we describe the design of novel amphiphilic pentablock copolymer (PBC) adjuvants that exhibit high biocompatibility and reversible pH- and temperature-sensitive micelle formation. We demonstrate improved humoral immunity in mice in response to single-dose immunization with PBC micelle adjuvants compared with soluble antigen alone. With the motive of exploring the mechanism of action of these PBC micelles, we studied intracellular trafficking of these PBC micelles with a model antigen and demonstrated that the PBC micelles associate with the antigen and enhance its cytosolic delivery to antigen-presenting cells. We posit that these PBC micelles operate via immune-enhancing mechanisms that are different from that of traditional Toll-like receptor activating adjuvants. The metabolic profile of antigen-presenting cells stimulated with traditional adjuvants and the PBC micelles also suggests distinct mechanisms of action. A key finding from this study is the low production of nitric oxide and reactive oxygen species by antigen-presenting cells when stimulated by PBC micelle adjuvants in sharp contrast to TLR adjuvants. Together, these studies provide a basis for rationally developing novel vaccine adjuvants that are safe, that induce low inflammation, and that can efficiently deliver antigen to the cytosol.
Collapse
Affiliation(s)
- Sujata Senapati
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Ross J. Darling
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Darren Loh
- Department of Chemical and Biological Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ian C. Schneider
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Michael J. Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Surya K. Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
27
|
Oldenkamp HF, Vela Ramirez JE, Peppas NA. Re-evaluating the importance of carbohydrates as regenerative biomaterials. Regen Biomater 2019; 6:1-12. [PMID: 30740237 PMCID: PMC6362819 DOI: 10.1093/rb/rby023] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/20/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- Heidi F Oldenkamp
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Julia E Vela Ramirez
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
28
|
Guerra-Maupome M, Palmer MV, McGill JL, Sacco RE. Utility of the Neonatal Calf Model for Testing Vaccines and Intervention Strategies for Use against Human RSV Infection. Vaccines (Basel) 2019; 7:vaccines7010007. [PMID: 30626099 PMCID: PMC6466205 DOI: 10.3390/vaccines7010007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/26/2018] [Accepted: 01/04/2019] [Indexed: 01/23/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a significant cause of pediatric respiratory tract infections. It is estimated that two-thirds of infants are infected with RSV during the first year of life and it is one of the leading causes of death in this age group worldwide. Similarly, bovine RSV is a primary viral pathogen in cases of pneumonia in young calves and plays a significant role in bovine respiratory disease complex. Importantly, naturally occurring infection of calves with bovine RSV shares many features in common with human RSV infection. Herein, we update our current understanding of RSV infection in cattle, with particular focus on similarities between the calf and human infection, and the recent reports in which the neonatal calf has been employed for the development and testing of vaccines and therapeutics which may be applied to hRSV infection in humans.
Collapse
Affiliation(s)
- Mariana Guerra-Maupome
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Mitchell V Palmer
- Infectious Bacterial Diseases of Livestock Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50010, USA.
| | - Jodi L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011, USA.
| | - Randy E Sacco
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA 50010, USA.
| |
Collapse
|
29
|
Nanotherapeutic provides dose sparing and improved antimicrobial activity against Brucella melitensis infections. J Control Release 2019; 294:288-297. [DOI: 10.1016/j.jconrel.2018.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 11/19/2022]
|
30
|
Rajput MKS, Kesharwani SS, Kumar S, Muley P, Narisetty S, Tummala H. Dendritic Cell-Targeted Nanovaccine Delivery System Prepared with an Immune-Active Polymer. ACS APPLIED MATERIALS & INTERFACES 2018; 10:27589-27602. [PMID: 30048112 DOI: 10.1021/acsami.8b02019] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Targeting dendritic cells (DCs), either ex vivo (Ex. Sipuleucel-T) or in vivo, for stimulating cellular immunity has been a leading approach for cancer vaccines. We have rationally engineered a nanoparticle (NP)-based delivery system for vaccines (InAc-NPs) using inulin acetate (InAc) as the polymer to target DCs. The material and the antigen-encapsulated InAc-NPs (∼190 nm in diameter) were characterized for their physicochemical properties. As a potent vaccine adjuvant, InAc-NPs activated TLR4 on multiple immune cells, including DCs and primary swine and human cells, to secrete various cytokines as detected by enzyme-linked immunosorbent assay and quantitative polymerase chain reaction. In addition, InAc-NPs promoted the maturation of DCs as observed by a decreased phagocytic ability and enhanced capability to activate various maturation markers (MHC-I, MHC-II, CD40, and CD80) quantified using flow cytometry. In mice, the InAc-NPs produced strong serum antibody titers (total IgG, IgG1, and IgG2a) against the encapsulated antigen (ovalbumin) similar to complete Freund's adjuvant. Additionally, as a dose-sparing delivery system, antigen delivered through InAc-NPs generated higher antibody titers (IgG1, 1.57 times; IgG-total, 1.66 times; and IgG2a, 29.8 times) even at 100 times less antigen dose. High amounts of cytokines representing both humoral (IL4 and IL10) and cell-mediated (IL2 and IFN-γ) immunities were secreted from splenocytes of mice immunized with InAc-NPs. Importantly, InAc-NPs provided complete protection in 100% of the vaccinated mice from metastasis of intravenously injected melanoma cells (B16-F10) to lungs. In addition, the InAc-NPs were cleared from the injection site within 30 h of injection (in vivo imaging) and displayed no toxicity at the injection site (histology). The current study demonstrates that the multifunctional InAc-based nanovaccine delivery system has potential applications in cancer immunotherapy and delivering vaccines against various infectious diseases.
Collapse
Affiliation(s)
- Mrigendra K S Rajput
- Department of Pharmaceutical Sciences , South Dakota State University , Box 2202C, Brookings , South Dakota 57007 , United States
- Department of Biological Sciences , Arkansas Tech University , Russellville , Arkansas 72801 , United States
| | - Siddharth S Kesharwani
- Department of Pharmaceutical Sciences , South Dakota State University , Box 2202C, Brookings , South Dakota 57007 , United States
| | - Sunny Kumar
- Department of Pharmaceutical Sciences , South Dakota State University , Box 2202C, Brookings , South Dakota 57007 , United States
| | - Pratik Muley
- Department of Pharmaceutical Sciences , South Dakota State University , Box 2202C, Brookings , South Dakota 57007 , United States
| | - Susmitha Narisetty
- Department of Pharmaceutical Sciences , South Dakota State University , Box 2202C, Brookings , South Dakota 57007 , United States
| | - Hemachand Tummala
- Department of Pharmaceutical Sciences , South Dakota State University , Box 2202C, Brookings , South Dakota 57007 , United States
| |
Collapse
|
31
|
Goodman JT, Mullis AS, Dunshee L, Mitra A, Narasimhan B. Automated High-Throughput Synthesis of Protein-Loaded Polyanhydride Nanoparticle Libraries. ACS COMBINATORIAL SCIENCE 2018; 20:298-307. [PMID: 29617113 DOI: 10.1021/acscombsci.8b00008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The development of high-throughput techniques and combinatorial libraries can facilitate rapid synthesis and screening of biomaterial-based nanocarriers for drug and vaccine delivery. This study describes a high-throughput method using an automated robot for synthesizing polyanhydride nanoparticles encapsulating proteins. Polyanhydrides are a class of safe and biodegradable polymers that have been widely used as drug and vaccine delivery vehicles. The robot contains a multiplexed homogenizer and has the capacity to handle parallel streams of monomer or polymer solutions to synthesize polymers and/or nanoparticles. Copolymer libraries were synthesized using the monomers sebacic acid, 1,6-bis( p-carboxyphenoxy)hexane, and 1,8-bis( p-carboxyphenoxy)-3,6-dioxactane and compared to conventionally synthesized copolymers. Nanoparticle libraries of varying copolymer compositions encapsulating the model antigen ovalbumin were synthesized using flash nanoprecipitation. The amount of the surfactant Span 80 was varied to test its effect on protein encapsulation efficiency as well as antigen release kinetics. It was observed that, although the amount of surfactant did not significantly affect protein release rate, its presence enhanced protein encapsulation efficiency. Protein burst and release kinetics from conventionally and combinatorially synthesized nanoparticles were similar even though particles synthesized using the high-throughput technique were smaller. Finally, it was demonstrated that the high-throughput method could be adapted to functionalize the surface of particle libraries to aid in the design and screening of targeted drug and vaccine delivery systems. These results suggest that the new high-throughput method is a viable alternative to conventional methods for synthesizing and screening protein and vaccine delivery vehicles.
Collapse
Affiliation(s)
- Jonathan T. Goodman
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Adam S. Mullis
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Lucas Dunshee
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Akash Mitra
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
32
|
Mullis AS, Schlichtmann BW, Narasimhan B, Cademartiri R, Mallapragada SK. Ligand-cascading nano-delivery devices to enable multiscale targeting of anti-neurodegenerative therapeutics. Biomed Mater 2018; 13:034102. [DOI: 10.1088/1748-605x/aaa778] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
33
|
Efficacy of mucosal polyanhydride nanovaccine against respiratory syncytial virus infection in the neonatal calf. Sci Rep 2018; 8:3021. [PMID: 29445124 PMCID: PMC5813012 DOI: 10.1038/s41598-018-21292-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) is a leading cause of severe acute lower respiratory tract infection in infants and children worldwide. Bovine RSV (BRSV) is closely related to HRSV and a significant cause of morbidity in young cattle. BRSV infection in calves displays many similarities to RSV infection in humans, including similar age dependency and disease pathogenesis. Polyanhydride nanoparticle-based vaccines (i.e., nanovaccines) have shown promise as adjuvants and vaccine delivery vehicles due to their ability to promote enhanced immunogenicity through the route of administration, provide sustained antigen exposure, and induce both antibody- and cell-mediated immunity. Here, we developed a novel, mucosal nanovaccine that encapsulates the post-fusion F and G glycoproteins from BRSV into polyanhydride nanoparticles and determined the efficacy of the vaccine against RSV infection using a neonatal calf model. Calves receiving the BRSV-F/G nanovaccine exhibited reduced pathology in the lungs, reduced viral burden, and decreased virus shedding compared to unvaccinated control calves, which correlated with BRSV-specific immune responses in the respiratory tract and peripheral blood. Our results indicate that the BRSV-F/G nanovaccine is highly immunogenic and, with optimization, has the potential to significantly reduce the disease burden associated with RSV infection in both humans and animals.
Collapse
|
34
|
Cheng H, Chawla A, Yang Y, Li Y, Zhang J, Jang HL, Khademhosseini A. Development of nanomaterials for bone-targeted drug delivery. Drug Discov Today 2017; 22:1336-1350. [PMID: 28487069 PMCID: PMC5644493 DOI: 10.1016/j.drudis.2017.04.021] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/15/2022]
Abstract
Bone is one of the major organs of the human body; it supports and protects other organs, produces blood cells, stores minerals, and regulates hormones. Therefore, disorders in bone can cause serious morbidity, complications, or mortality of patients. However, despite the significant occurrence of bone diseases, such as osteoarthritis (OA), osteoporosis (OP), non-union bone defects, bone cancer, and myeloma-related bone disease, their effective treatments remain a challenge. In this review, we highlight recent progress in the development of nanotechnology-based drug delivery for bone treatment, based on its improved delivery efficiency and safety. We summarize the most commonly used nanomaterials for bone drug delivery. We then discuss the targeting strategies of these nanomaterials to the diseased sites of bone tissue. We also highlight nanotechnology-based drug delivery to bone cells and subcellular organelles. We envision that nanotechnology-based drug delivery will serve as a powerful tool for developing treatments for currently incurable bone diseases.
Collapse
Affiliation(s)
- Hao Cheng
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02139, USA; Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Orthopaedic Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Aditya Chawla
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02139, USA; Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Yafeng Yang
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02139, USA; Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuxiao Li
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02139, USA; Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jin Zhang
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02139, USA; Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hae Lin Jang
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02139, USA; Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| | - Ali Khademhosseini
- Division of Biomedical Engineering, Department of Medicine, Biomaterials Innovation Research Center, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02139, USA; Division of Health Sciences & Technology, Harvard-Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Bioindustrial Technologies, College of Animal Bioscience & Technology, Konkuk University, Seoul 143-701, Republic of Korea; Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia.
| |
Collapse
|
35
|
Xue B, Wang W, Qin JJ, Nijampatnam B, Murugesan S, Kozlovskaya V, Zhang R, Velu SE, Kharlampieva E. Highly efficient delivery of potent anticancer iminoquinone derivative by multilayer hydrogel cubes. Acta Biomater 2017; 58:386-398. [PMID: 28583901 PMCID: PMC5736006 DOI: 10.1016/j.actbio.2017.06.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/15/2017] [Accepted: 06/02/2017] [Indexed: 01/04/2023]
Abstract
We report a novel delivery platform for a highly potent anticancer drug, 7-(benzylamino)-3,4-dihydro-pyrrolo[4,3,2-de]quinolin-8(1H)-one (BA-TPQ), using pH- and redox-sensitive poly(methacrylic acid) (PMAA) hydrogel cubes of micrometer size as the encapsulating matrix. The hydrogels are obtained upon cross-linking PMAA with cystamine in PMAA/poly(N-vinylpyrrolidone) multilayers assembled within mesoporous sacrificial templates. The BA-TPQ-loaded hydrogels maintain their cubical shape and pH-sensitivity after lyophilization, which is advantageous for long-term storage. Conversely, the particles degrade in vitro in the presence of glutathione (5mM) providing 80% drug release within 24h. Encapsulating BA-TPQ into hydrogels significantly increases its transport via Caco-2 cell monolayers used as a model for oral delivery where the apparent permeability of BA-TPQ-hydrogel cubes was∼2-fold higher than that of BA-TPQ. BA-TPQ-hydrogel cubes exhibit better anticancer activity against HepG2 (IC50=0.52µg/mL) and Huh7 (IC50=0.29µg/mL) hepatoma cells with a 40% decrease in the IC50 compared to the non-encapsulated drug. Remarkably, non-malignant liver cells have a lower sensitivity to BA-TPQ-hydrogel cubes with 2-fold increased IC50 values compared to those of cancer cells. In addition, encapsulating BA-TPQ in the hydrogels amplifies the potency of the drug via down-regulation of MDM2 oncogenic protein and upregulation of p53 (a tumor suppressor) and p21 (cell proliferation suppressor) expression in HepG2 liver cancer cells. Moreover, enhanced inhibition of MDM2 protein expression by BA-TPQ-hydrogel cubes is independent of p53 status in Huh7 cells. This drug delivery platform of non-spherical shape provides a facile method for encapsulation of hydrophobic drugs and can facilitate the enhanced efficacy of BA-TPQ for liver cancer therapy. STATEMENT OF SIGNIFICANCE Many potent anticancer drugs are hydrophobic and lack tumor selectivity, which limits their application in cancer therapy. Although cubical hydrogels of poly(methacrylic acid) exhibit excellent biocompatibility and versatility, they have not been investigated for hydrophobic drug delivery due to poor mechanical stability and incompatibility between hydrophobic drugs and a hydrophilic hydrogel network. In this study, we provide a facile method to prepare a multilayer hydrogel-based platform with controlled nanostructure, cubical shape and redox-responsiveness for delivery of highly potent anticancer therapeutics, hydrophobic BA-TPQ. The BA-TPQ-hydrogel cubes have exceptional structural stability upon lyophilization which is advantageous for a long-term storage. The greatly enhanced trans-epithelial permeability and amplified anti-tumor activity of BA-TPQ are achieved by encapsulation in these hydrogel cubes. Furthermore, the anticancer BA-TPQ-hydrogel platform retains the selective activity of BA-TPQ to hepatocellular carcinoma cells. Overall, the produced BA-TPQ-hydrogel cubes demonstrate a high potential for clinical liver cancer therapy.
Collapse
Affiliation(s)
- Bing Xue
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States
| | - Jiang-Jiang Qin
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States
| | - Bhavitavya Nijampatnam
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Srinivasan Murugesan
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Veronika Kozlovskaya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Ruiwen Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, United States.
| | - Sadanandan E Velu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294-3300, United States.
| | - Eugenia Kharlampieva
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Center of Nanoscale Materials and Biointegration, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
36
|
Zerbe K, Moehle K, Robinson JA. Protein Epitope Mimetics: From New Antibiotics to Supramolecular Synthetic Vaccines. Acc Chem Res 2017; 50:1323-1331. [PMID: 28570824 DOI: 10.1021/acs.accounts.7b00129] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Protein epitope mimetics provide powerful tools to study biomolecular recognition in many areas of chemical biology. They may also provide access to new biologically active molecules and potentially to new classes of drug and vaccine candidates. Here we highlight approaches for the design of folded, structurally defined epitope mimetics, by incorporating backbone and side chains of hot residues onto a stable constrained scaffold. Using robust synthetic methods, the structural, biological, and physical properties of epitope mimetics can be optimized, by variation of both side chain and backbone chemistry. To illustrate the potential of protein epitope mimetics in medicinal chemistry and biotechnology, we present studies in two areas of infectology; the discovery of new antibiotics targeting essential outer membrane (OM) proteins in Gram-negative bacteria and the design of supramolecular synthetic vaccines. The discovery of new antibiotics with novel mechanisms of action, in particular to combat infections caused by Gram-negative pathogens, represents a major challenge in medicinal chemistry. We were inspired by naturally occurring cationic antimicrobial peptides to design structurally related peptidomimetics and to optimize their antimicrobial properties through library synthesis and screening. Through these efforts, we could show that antimicrobial β-hairpin mimetics may have structures and properties that facilitate interactions with essential bacterial β-barrel OM proteins. One recently discovered family of antimicrobial peptidomimetics targets the β-barrel protein LptD in Pseudomonas spp. This protein plays a key role in lipopolysaccaride (LPS) transport to the cell surface during OM biogenesis. Through a highly selective interaction with LptD, the peptidomimetic blocks LPS transport, resulting in nanomolar antimicrobial activity against the important human pathogen P. aeruginosa. Epitope mimetics may also have great potential in the field of vaccinology, where structural information on complexes between neutralizing antibodies and their cognate epitopes can be taken as a starting point for B cell epitope mimetic design. In order to generate potent immune responses, an effective method of delivering epitope mimetics to relevant cells and tissues in the immune system is also required. For this, engineered synthetic nanoparticles (synthetic virus-like particles, SVLPs) prepared using supramolecular chemistry can be designed with optimal surface properties for efficient dendritic cell-mediated delivery of folded B-cell and linear T-cell epitopes, along with ligands for pattern recognition receptors, into lymphoid tissues. In this way, multivalent display of the epitope mimetics occurs over the surface of the nanoparticle, suitable for cross-linking B cell receptors. In this highly immunogenic format, strong epitope-specific humoral immune responses can be elicited that target infections caused by pathogenic microorganisms. Other potential applications of epitope mimetics in next-generation therapeutics are also discussed.
Collapse
Affiliation(s)
- Katja Zerbe
- Chemistry Department, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Kerstin Moehle
- Chemistry Department, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - John A. Robinson
- Chemistry Department, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
37
|
Vela Ramirez JE, Sharpe LA, Peppas NA. Current state and challenges in developing oral vaccines. Adv Drug Deliv Rev 2017; 114:116-131. [PMID: 28438674 PMCID: PMC6132247 DOI: 10.1016/j.addr.2017.04.008] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/10/2017] [Accepted: 04/19/2017] [Indexed: 02/06/2023]
Abstract
While vaccination remains the most cost effective strategy for disease prevention, communicable diseases persist as the second leading cause of death worldwide. There is a need to design safe, novel vaccine delivery methods to protect against unaddressed and emerging diseases. Development of vaccines administered orally is preferable to traditional injection-based formulations for numerous reasons including improved safety and compliance, and easier manufacturing and administration. Additionally, the oral route enables stimulation of humoral and cellular immune responses at both systemic and mucosal sites to establish broader and long-lasting protection. However, oral delivery is challenging, requiring formulations to overcome the harsh gastrointestinal (GI) environment and avoid tolerance induction to achieve effective protection. Here we address the rationale for oral vaccines, including key biological and physicochemical considerations for next-generation oral vaccine design.
Collapse
Affiliation(s)
- Julia E Vela Ramirez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Lindsey A Sharpe
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA; McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA; Division of Pharmaceutics, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
38
|
Malik T, Chauhan G, Rath G, Murthy RSR, Goyal AK. "Fusion and binding inhibition" key target for HIV-1 treatment and pre-exposure prophylaxis: targets, drug delivery and nanotechnology approaches. Drug Deliv 2017; 24:608-621. [PMID: 28240046 PMCID: PMC8241151 DOI: 10.1080/10717544.2016.1228717] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
More than 35 million people are living with HIV worldwide with approximately 2.3 million new infections per year. Cascade of events (cell entry, virus replication, assembly and release of newly formed virions) is involved in the HIV-1 transmission process. Every single step offers a potential therapeutic strategy to halt this progression and HIV fusion into the human host cell is one such stage. Controlling the initial event of HIV-1 transmission is the best way to control its dissemination especially when prophylaxis is concerned. Action is required either on the HIV’s or host’s cell surface which is logically more rational when compared with other intracellular acting moieties. Aim of this manuscript is to detail the significance and current strategies to halt this initial step, thus blocking the entry of HIV-1 for further infection. Both HIV-1 and the possible host cell’s receptors/co-receptors are under focus while specifying the targets available for inhibiting this fusion. Current and under investigation moieties are categorized based on their versatile mechanisms. Advanced drug delivery and nanotechnology approaches present a key tool to exploit the therapeutic potential in a boosted way. Current drug delivery and the impact of nanotechnology in potentiating this strategy are detailed.
Collapse
Affiliation(s)
- Tanushree Malik
- a DBT Lab, Indo Soviet Friendship College of Pharmacy , Moga , India and
| | - Gaurav Chauhan
- a DBT Lab, Indo Soviet Friendship College of Pharmacy , Moga , India and.,b Centre for Nanosciences, Department of Chemical Engineering, Indian Institute of Technology Kanpur , Kanpur , India
| | - Goutam Rath
- a DBT Lab, Indo Soviet Friendship College of Pharmacy , Moga , India and
| | - R S R Murthy
- a DBT Lab, Indo Soviet Friendship College of Pharmacy , Moga , India and
| | - Amit K Goyal
- a DBT Lab, Indo Soviet Friendship College of Pharmacy , Moga , India and
| |
Collapse
|
39
|
Brenza TM, Ghaisas S, Ramirez JEV, Harischandra D, Anantharam V, Kalyanaraman B, Kanthasamy AG, Narasimhan B. Neuronal protection against oxidative insult by polyanhydride nanoparticle-based mitochondria-targeted antioxidant therapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:809-820. [PMID: 27771430 DOI: 10.1016/j.nano.2016.10.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/30/2016] [Accepted: 10/10/2016] [Indexed: 12/17/2022]
Abstract
A progressive loss of neuronal structure and function is a signature of many neurodegenerative conditions including chronic traumatic encephalopathy, Parkinson's, Huntington's and Alzheimer's diseases. Mitochondrial dysfunction and oxidative and nitrative stress have been implicated as key pathological mechanisms underlying the neurodegenerative processes. However, current therapeutic approaches targeting oxidative damage are ineffective in preventing the progression of neurodegeneration. Mitochondria-targeted antioxidants were recently shown to alleviate oxidative damage. In this work, we investigated the delivery of biodegradable polyanhydride nanoparticles containing the mitochondria-targeted antioxidant apocynin to neuronal cells and the ability of the nano-formulation to protect cells against oxidative stress. The nano-formulated mitochondria-targeted apocynin provided excellent protection against oxidative stress-induced mitochondrial dysfunction and neuronal damage in a dopaminergic neuronal cell line, mouse primary cortical neurons, and a human mesencephalic cell line. Collectively, our results demonstrate that nano-formulated mitochondria-targeted apocynin may offer improved efficacy of mitochondria-targeted antioxidants to treat neurodegenerative disease.
Collapse
Affiliation(s)
- Timothy M Brenza
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Shivani Ghaisas
- Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Julia E Vela Ramirez
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | | | | | | | | | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA.
| |
Collapse
|
40
|
Kharrat N, Belmabrouk S, Abdelhedi R, Benmarzoug R, Assidi M, Al Qahtani MH, Rebai A. Screening for clusters of charge in human virus proteomes. BMC Genomics 2016; 17:758. [PMID: 27766959 PMCID: PMC5073957 DOI: 10.1186/s12864-016-3086-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background The identification of charge clusters (runs of charged residues) in proteins and their mapping within the protein structure sequence is an important step toward a comprehensive analysis of how these particular motifs mediate, via electrostatic interactions, various molecular processes such as protein sorting, translocation, docking, orientation and binding to DNA and to other proteins. Few algorithms that specifically identify these charge clusters have been designed and described in the literature. In this study, 197 distinctive human viral proteomes were screened for the occurrence of charge clusters (CC) using a new computational approach. Results Three hundred and seventy three CC have been identified within the 2549 viral protein sequences screened. The number of protein sequences that are CC-free is 2176 (85.3 %) while 150 and 180 proteins contained positive charge (PCC) and negative charge clusters (NCC), respectively. The NCCs (211 detected) were more prevalent than PCC (162). PCC-containing proteins are significantly longer than those having NCCs (p = 2.10-16). The most prevalent virus families having PCC and NCC were Herpesviridae followed by Papillomaviridae. However, the single-strand RNA group has in average three times more NCC than PCC. According to the functional domain classification, a significant difference in distribution was observed between PCC and NCC (p = 2. 10−8) with the occurrence of NCCs being more frequent in C-terminal region while PCC more often fall within functional domains. Only 29 proteins sequences contained both NCC and PCC. Moreover, 101 NCC were conserved in 84 proteins while only 62 PCC were conserved in 60 protein sequences. To understand the mechanism by which the membrane translocation functionalities are embedded in viral proteins, we screened our PCC for sequences corresponding to cell-penetrating peptides (CPPs) using two online databases: CellPPd and CPPpred. We found that all our PCCs, having length varying from 7 to 30 amino-acids were predicted as CPPs. Experimental validation is required to improve our understanding of the role of these PCCs in viral infection process. Conclusions Screening distinctive cluster charges in viral proteomes suggested a functional role of these protein regions and might provide potential clues to improve the current understanding of viral diseases in order to tailor better preventive and therapeutic approaches. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3086-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Najla Kharrat
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia.
| | - Sabrine Belmabrouk
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia
| | - Rania Abdelhedi
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia
| | - Riadh Benmarzoug
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia
| | - Mourad Assidi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Center of Innovation in Personalized Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed H Al Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Rebai
- Centre of Biotechnology of Sfax, Laboratory of Molecular and Cellular Screening Processes, Bioinformatics Group, PO. Box:1177, 3018, Sfax, Tunisia
| |
Collapse
|
41
|
Vela Ramirez JE, Boggiatto PM, Wannemuehler MJ, Narasimhan B. Polyanhydride Nanoparticle Interactions with Host Serum Proteins and Their Effects on Bone Marrow Derived Macrophage Activation. ACS Biomater Sci Eng 2016; 3:160-168. [PMID: 33450792 DOI: 10.1021/acsbiomaterials.6b00394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
An in-depth understanding of the interactions of vaccine delivery vehicles with antigen presenting cells is important for tailoring optimal adjuvant properties. Polymeric nanoparticles have been widely studied as adjuvants and delivery vehicles; however, there is little information regarding the effect of serum protein adsorption onto biomaterials and the effect of this adsorption upon interactions with antigen presenting cells. The current studies analyzed effects of polyanhydride chemistry on serum adsorption to nanoparticles with respect to their uptake by and activation of bone marrow-derived macrophages. Differential effects of serum adsorption based on nanoparticle chemistry were shown to enhance (for 1,6-bis(p-carboxyphenoxy)hexane and sebacic anhydride-based) or reduce (for 1,6-bis(p-carboxyphenoxy)hexane and 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane-based) nanoparticle uptake. The observed complex interdependence between nanoparticle chemistry and serum protein adsorption on macrophage activation provided insights that will facilitate the rational design of single-dose nanovaccines developed to induce robust immune responses.
Collapse
Affiliation(s)
- Julia E Vela Ramirez
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Paola M Boggiatto
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|