1
|
Sadr S, Poorjafari Jafroodi P, Haratizadeh MJ, Ghasemi Z, Borji H, Hajjafari A. Current status of nano-vaccinology in veterinary medicine science. Vet Med Sci 2023; 9:2294-2308. [PMID: 37487030 PMCID: PMC10508510 DOI: 10.1002/vms3.1221] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/11/2023] [Accepted: 07/14/2023] [Indexed: 07/26/2023] Open
Abstract
Vaccination programmes provide a safe, effective and cost-efficient strategy for maintaining population health. In veterinary medicine, vaccination not only reduces disease within animal populations but also serves to enhance public health by targeting zoonoses. Nevertheless, for many pathogens, an effective vaccine remains elusive. Recently, nanovaccines have proved to be successful for various infectious and non-infectious diseases of animals. These novel technologies, such as virus-like particles, self-assembling proteins, polymeric nanoparticles, liposomes and virosomes, offer great potential for solving many of the vaccine production challenges. Their benefits include low immunotoxicity, antigen stability, enhanced immunogenicity, flexibility sustained release and the ability to evoke both humoral and cellular immune responses. Nanovaccines are more efficient than traditional vaccines due to ease of control and plasticity in their physio-chemical properties. They use a highly targeted immunological approach which can provide strong and long-lasting immunity. This article reviews the currently available nanovaccine technology and considers its utility for both infectious diseases and non-infectious diseases such as auto-immunity and cancer. Future research opportunities and application challenges from bench to clinical usage are also discussed.
Collapse
Affiliation(s)
- Soheil Sadr
- Department of Clinical SciencesFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | | | | | - Zahra Ghasemi
- Department of Clinical SciencesFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | - Hassan Borji
- Department of PathobiologyFaculty of Veterinary MedicineFerdowsi University of MashhadMashhadIran
| | - Ashkan Hajjafari
- Department of PathobiologyFaculty of Veterinary MedicineIslamic Azad University, Science and Research BranchTehranIran
| |
Collapse
|
2
|
Maina TW, Grego EA, Broderick S, Sacco RE, Narasimhan B, McGill JL. Immunization with a mucosal, post-fusion F/G protein-based polyanhydride nanovaccine protects neonatal calves against BRSV infection. Front Immunol 2023; 14:1186184. [PMID: 37359514 PMCID: PMC10289034 DOI: 10.3389/fimmu.2023.1186184] [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: 03/14/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Human respiratory syncytial virus (HRSV) is a leading cause of death in young children and there are no FDA approved vaccines. Bovine RSV (BRSV) is antigenically similar to HRSV, and the neonatal calf model is useful for evaluation of HRSV vaccines. Here, we determined the efficacy of a polyanhydride-based nanovaccine encapsulating the BRSV post-fusion F and G glycoproteins and CpG, delivered prime-boost via heterologous (intranasal/subcutaneous) or homologous (intranasal/intranasal) immunization in the calf model. We compared the performance of the nanovaccine regimens to a modified-live BRSV vaccine, and to non-vaccinated calves. Calves receiving nanovaccine via either prime-boost regimen exhibited clinical and virological protection compared to non-vaccinated calves. The heterologous nanovaccine regimen induced both virus-specific cellular immunity and mucosal IgA, and induced similar clinical, virological and pathological protection as the commercial modified-live vaccine. Principal component analysis identified BRSV-specific humoral and cellular responses as important correlates of protection. The BRSV-F/G CpG nanovaccine is a promising candidate vaccine to reduce RSV disease burden in humans and animals.
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
| | - Scott Broderick
- Department of Materials Design and Innovation, University at Buffalo, Buffalo, NY, United States
| | - Randy E. Sacco
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture (USDA), 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
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
- Nanovaccine Institute, Iowa State University, Ames, IA, United States
| |
Collapse
|
3
|
Phanse Y, Puttamreddy S, Loy D, Ramirez JV, Ross KA, Alvarez-Castro I, Mogler M, Broderick S, Rajan K, Narasimhan B, Bartholomay LC. RNA Nanovaccine Protects against White Spot Syndrome Virus in Shrimp. Vaccines (Basel) 2022; 10:vaccines10091428. [PMID: 36146509 PMCID: PMC9504209 DOI: 10.3390/vaccines10091428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
In the last 15 years, crustacean fisheries have experienced billions of dollars in economic losses, primarily due to viral diseases caused by such pathogens as white spot syndrome virus (WSSV) in the Pacific white shrimp Litopenaeus vannamei and Asian tiger shrimp Penaeus monodon. To date, no effective measures are available to prevent or control disease outbreaks in these animals, despite their economic importance. Recently, double-stranded RNA-based vaccines have been shown to provide specific and robust protection against WSSV infection in cultured shrimp. However, the limited stability of double-stranded RNA is the most significant hurdle for the field application of these vaccines with respect to delivery within an aquatic system. Polyanhydride nanoparticles have been successfully used for the encapsulation and release of vaccine antigens. We have developed a double-stranded RNA-based nanovaccine for use in shrimp disease control with emphasis on the Pacific white shrimp L. vannamei. Nanoparticles based on copolymers of sebacic acid, 1,6-bis(p-carboxyphenoxy)hexane, and 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane exhibited excellent safety profiles, as measured by shrimp survival and histological evaluation. Furthermore, the nanoparticles localized to tissue target replication sites for WSSV and persisted through 28 days postadministration. Finally, the nanovaccine provided ~80% protection in a lethal WSSV challenge model. This study demonstrates the exciting potential of a safe, effective, and field-applicable RNA nanovaccine that can be rationally designed against infectious diseases affecting aquaculture.
Collapse
Affiliation(s)
- Yashdeep Phanse
- Department of Entomology, Iowa State University, Ames, IA 50011, USA
- Pan Genome Systems, Madison, WI 53719, USA
| | - Supraja Puttamreddy
- Department of Entomology, Iowa State University, Ames, IA 50011, USA
- Merck Animal Health, Ames, IA 50010, USA
| | - Duan Loy
- Department of Entomology, Iowa State University, Ames, IA 50011, USA
- Veterinary Diagnostics Center, University of Nebraska Lincoln, Lincoln, NE 68583, USA
| | - Julia Vela Ramirez
- Department of Chemical and Biological Engineering, Nanovaccine Institute, Iowa State University, Ames, IA 50011, USA
| | - Kathleen A. Ross
- Department of Chemical and Biological Engineering, Nanovaccine Institute, Iowa State University, Ames, IA 50011, USA
| | | | - Mark Mogler
- Merck Animal Health, Ames, IA 50010, USA
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - Scott Broderick
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011, USA
- Department of Materials Design and Innovation, University at Buffalo, Buffalo, NY 14260, USA
| | - Krishna Rajan
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011, USA
- Department of Materials Design and Innovation, University at Buffalo, Buffalo, NY 14260, USA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Nanovaccine Institute, Iowa State University, Ames, IA 50011, USA
- Correspondence: (B.N.); (L.C.B.); Tel.: +1-515-294-8019 (B.N.); +1-608-890-1965 (L.C.B.)
| | - Lyric C. Bartholomay
- Department of Entomology, Iowa State University, Ames, IA 50011, USA
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
- Correspondence: (B.N.); (L.C.B.); Tel.: +1-515-294-8019 (B.N.); +1-608-890-1965 (L.C.B.)
| |
Collapse
|
4
|
Siddoway AC, Verhoeven D, Ross KA, Wannemuehler MJ, Mallapragada SK, Narasimhan B. Structural Stability and Antigenicity of Universal Equine H3N8 Hemagglutinin Trimer upon Release from Polyanhydride Nanoparticles and Pentablock Copolymer Hydrogels. ACS Biomater Sci Eng 2022; 8:2500-2507. [PMID: 35604784 DOI: 10.1021/acsbiomaterials.2c00219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Seasonal influenza A virus infections present substantial costs to both health and economic resources each year. Current seasonal influenza vaccines provide suboptimal protection and require annual reformulation to match circulating strains. In this work, a recombinant equine H3N8 hemagglutinin trimer (rH33) known to generate cross-protective antibodies and protect animals against sublethal, heterologous virus challenge was used as a candidate vaccine antigen. Nanoadjuvants such as polyanhydride nanoparticles and pentablock copolymer hydrogels have been shown to be effective adjuvants, inducing both rapid and long-lived protective immunity against influenza A virus. In this work, polyanhydride nanoparticles and pentablock copolymer hydrogels were used to provide sustained release of the novel rH33 while also facilitating the retention of its structure and antigenicity. These studies lay the groundwork for the development of a novel universal influenza A virus nanovaccine by combining the equine H3N8 rH33 and polymeric nanoadjuvant platforms.
Collapse
Affiliation(s)
- Alaric C Siddoway
- Department of Chemical & Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - David Verhoeven
- Department of Veterinary Microbiology & Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Ames, Iowa 50011, United States
| | | | - Michael J Wannemuehler
- Department of Veterinary Microbiology & Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Ames, Iowa 50011, United States
| | - Surya K Mallapragada
- Department of Chemical & Biological Engineering, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical & Biological Engineering, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Ames, Iowa 50011, United States
| |
Collapse
|
5
|
Ryabchevskaya EM, Granovskiy DL, Evtushenko EA, Ivanov PA, Kondakova OA, Nikitin NA, Karpova OV. Designing Stable Bacillus anthracis Antigens with a View to Recombinant Anthrax Vaccine Development. Pharmaceutics 2022; 14:pharmaceutics14040806. [PMID: 35456639 PMCID: PMC9025368 DOI: 10.3390/pharmaceutics14040806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
Anthrax is a disease caused by Bacillus anthracis that affects mammals, including humans. Recombinant B. anthracis protective antigen (rPA) is the most common basis for modern anthrax vaccine candidates. However, this protein is characterised by low stability due to proteolysis and deamidation. Here, for the first time, two modification variants leading to full-size rPA stabilisation have been implemented simultaneously, through deamidation-prone asparagine residues substitution and by inactivation of proteolysis sites. Obtained modified rPA (rPA83m) has been demonstrated to be stable in various temperature conditions. Additionally, rPA1+2 containing PA domains I and II and rPA3+4 containing domains III and IV, including the same modifications, have been shown to be stable as well. These antigens can serve as the basis for a vaccine, since the protective properties of PA can be attributed to individual PA domains. The stability of each of three modified anthrax antigens has been considerably improved in compositions with tobacco mosaic virus-based spherical particles (SPs). rPA1+2/rPA3+4/rPA83m in compositions with SPs have maintained their antigenic specificity even after 40 days of incubation at +37 °C. Considering previously proven adjuvant properties and safety of SPs, their compositions with rPA83m/rPA1+2/rPA3+4 in any combinations might be suitable as a basis for new-generation anthrax vaccines.
Collapse
|
6
|
Akache B, Stark FC, Agbayani G, Renner TM, McCluskie MJ. Adjuvants: Engineering Protective Immune Responses in Human and Veterinary Vaccines. Methods Mol Biol 2022; 2412:179-231. [PMID: 34918246 DOI: 10.1007/978-1-0716-1892-9_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adjuvants are key components of many vaccines, used to enhance the level and breadth of the immune response to a target antigen, thereby enhancing protection from the associated disease. In recent years, advances in our understanding of the innate and adaptive immune systems have allowed for the development of a number of novel adjuvants with differing mechanisms of action. Herein, we review adjuvants currently approved for human and veterinary use, describing their use and proposed mechanisms of action. In addition, we will discuss additional promising adjuvants currently undergoing preclinical and/or clinical testing.
Collapse
Affiliation(s)
- Bassel Akache
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Felicity C Stark
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Gerard Agbayani
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Tyler M Renner
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada
| | - Michael J McCluskie
- Human Health Therapeutics, National Research Council Canada, Ottawa, ON, Canada.
| |
Collapse
|
7
|
Dassanayake RP, Atkinson BM, Mullis AS, Falkenberg SM, Nicholson EM, Casas E, Narasimhan B, Bearson SMD. Bovine NK-lysin peptides exert potent antimicrobial activity against multidrug-resistant Salmonella outbreak isolates. Sci Rep 2021; 11:19276. [PMID: 34588573 PMCID: PMC8481502 DOI: 10.1038/s41598-021-98860-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023] Open
Abstract
Multidrug-resistant (MDR) Salmonella is a threat to public health. Non-antibiotic therapies could serve as important countermeasures to control MDR Salmonella outbreaks. In this study, antimicrobial activity of cationic α-helical bovine NK-lysin-derived antimicrobial peptides was evaluated against MDR Salmonella outbreak isolates. NK2A and NK2B strongly inhibited MDR Salmonella growth while NK1 and NK2C showed minimum-to-no growth inhibition. Scrambled-NK2A, which is devoid of α-helicity but has the same net positive charge as NK2A, also failed to inhibit bacterial growth. Incubation of negatively charged MDR Salmonella with NK2A showed increased Zeta potential, indicating bacterial-peptide electrostatic attraction. Confocal and transmission electron microscopy studies revealed NK2A-mediated damage to MDR Salmonella membranes. LPS inhibited NK2A-mediated growth suppression in a dose-dependent response, suggesting irreversible NK2A-LPS binding. LPS-NK2A binding and bacterial membrane disruption was also confirmed via electron microscopy using gold nanoparticle-NK2A conjugates. Finally, NK2A-loaded polyanhydride nanoparticles showed sustained peptide delivery and anti-bacterial activity. Together, these findings indicate that NK2A α-helicity and positive charge are prerequisites for antimicrobial activity and that MDR Salmonella killing is mediated by direct interaction of NK2A with LPS and the inner membrane, leading to bacterial membrane permeabilization. With further optimization using nano-carriers, NK2A has the potential to become a potent anti-MDR Salmonella agent.
Collapse
Affiliation(s)
- Rohana P Dassanayake
- Agricultural Research Service, National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, USDA, Ames, IA, USA.
| | - Briony M Atkinson
- Agricultural Research Service, National Animal Disease Center, Food Safety and Enteric Pathogens Research Unit, USDA, Ames, IA, USA
| | - Adam S Mullis
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Shollie M Falkenberg
- Agricultural Research Service, National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, USDA, Ames, IA, USA
| | - Eric M Nicholson
- Virus and Prion Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Eduardo Casas
- Agricultural Research Service, National Animal Disease Center, Ruminant Diseases and Immunology Research Unit, USDA, Ames, IA, USA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering and Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Shawn M D Bearson
- Agricultural Research Service, National Animal Disease Center, Food Safety and Enteric Pathogens Research Unit, USDA, Ames, IA, USA.
| |
Collapse
|
8
|
Seyfoori A, Shokrollahi Barough M, Mokarram P, Ahmadi M, Mehrbod P, Sheidary A, Madrakian T, Kiumarsi M, Walsh T, McAlinden KD, Ghosh CC, Sharma P, Zeki AA, Ghavami S, Akbari M. Emerging Advances of Nanotechnology in Drug and Vaccine Delivery against Viral Associated Respiratory Infectious Diseases (VARID). Int J Mol Sci 2021; 22:6937. [PMID: 34203268 PMCID: PMC8269337 DOI: 10.3390/ijms22136937] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/19/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022] Open
Abstract
Viral-associated respiratory infectious diseases are one of the most prominent subsets of respiratory failures, known as viral respiratory infections (VRI). VRIs are proceeded by an infection caused by viruses infecting the respiratory system. For the past 100 years, viral associated respiratory epidemics have been the most common cause of infectious disease worldwide. Due to several drawbacks of the current anti-viral treatments, such as drug resistance generation and non-targeting of viral proteins, the development of novel nanotherapeutic or nano-vaccine strategies can be considered essential. Due to their specific physical and biological properties, nanoparticles hold promising opportunities for both anti-viral treatments and vaccines against viral infections. Besides the specific physiological properties of the respiratory system, there is a significant demand for utilizing nano-designs in the production of vaccines or antiviral agents for airway-localized administration. SARS-CoV-2, as an immediate example of respiratory viruses, is an enveloped, positive-sense, single-stranded RNA virus belonging to the coronaviridae family. COVID-19 can lead to acute respiratory distress syndrome, similarly to other members of the coronaviridae. Hence, reviewing the current and past emerging nanotechnology-based medications on similar respiratory viral diseases can identify pathways towards generating novel SARS-CoV-2 nanotherapeutics and/or nano-vaccines.
Collapse
Affiliation(s)
- Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Mahdieh Shokrollahi Barough
- Department of Immunology, Iran University of Medical Sciences, Tehran 1449614535, Iran;
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Pooneh Mokarram
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran;
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran 1316943551, Iran;
| | - Alireza Sheidary
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Mohammad Kiumarsi
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Tavia Walsh
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
| | - Kielan D. McAlinden
- Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia;
| | - Chandra C. Ghosh
- Roger Williams Medical Center, Immuno-Oncology Institute (Ix2), Providence, RI 02908, USA;
| | - Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Amir A. Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, U.C. Davis Lung Center, Davis School of Medicine, University of California, Davis, CA 95817, USA;
- Veterans Affairs Medical Center, Mather, CA 95817, USA
| | - Saeid Ghavami
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
9
|
Kelly SM, Larsen KR, Darling R, Petersen AC, Bellaire BH, Wannemuehler MJ, Narasimhan B. Single-dose combination nanovaccine induces both rapid and durable humoral immunity and toxin neutralizing antibody responses against Bacillus anthracis. Vaccine 2021; 39:3862-3870. [PMID: 34090702 DOI: 10.1016/j.vaccine.2021.05.077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/24/2021] [Accepted: 05/23/2021] [Indexed: 12/11/2022]
Abstract
Bacillus anthracis, the causative agent of anthrax, continues to be a prominent biological warfare and bioterrorism threat. Vaccination is likely to remain the most effective and user-friendly public health measure to counter this threat in the foreseeable future. The commercially available AVA BioThrax vaccine has a number of shortcomings where improvement would lead to a more practical and effective vaccine for use in the case of an exposure event. Identification of more effective adjuvants and novel delivery platforms is necessary to improve not only the effectiveness of the anthrax vaccine, but also enhance its shelf stability and ease-of-use. Polyanhydride particles have proven to be an effective platform at adjuvanting the vaccine-associated adaptive immune response as well as enhancing stability of encapsulated antigens. Another class of adjuvants, the STING pathway-targeting cyclic dinucleotides, have proven to be uniquely effective at inducing a beneficial inflammatory response that leads to the rapid induction of high titer antibodies post-vaccination capable of providing protection against bacterial pathogens. In this work, we evaluate the individual contributions of cyclic di-GMP (CDG), polyanhydride nanoparticles, and a combination thereof towards inducing neutralizing antibody (nAb) against the secreted protective antigen (PA) from B. anthracis. Our results show that the combination nanovaccine elicited rapid, high titer, and neutralizing IgG anti-PA antibody following single dose immunization that persisted for at least 108 DPI.
Collapse
Affiliation(s)
- Sean M Kelly
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States; Nanovaccine Institute, Ames, IA, United States
| | - Kristina R Larsen
- 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 Darling
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Andrew C Petersen
- 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, Ames, IA, United States
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States; Nanovaccine Institute, Ames, IA, United States.
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States; Nanovaccine Institute, Ames, IA, United States.
| |
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: 28] [Impact Index Per Article: 7.0] [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
|
Stephens LM, Ross KA, Waldstein KA, Legge KL, McLellan JS, Narasimhan B, Varga SM. Prefusion F-Based Polyanhydride Nanovaccine Induces Both Humoral and Cell-Mediated Immunity Resulting in Long-Lasting Protection against Respiratory Syncytial Virus. THE JOURNAL OF IMMUNOLOGY 2021; 206:2122-2134. [PMID: 33827894 DOI: 10.4049/jimmunol.2100018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/19/2021] [Indexed: 11/19/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection in both young children and in older adults. Despite the morbidity, mortality, and high economic burden caused by RSV worldwide, no licensed vaccine is currently available. We have developed a novel RSV vaccine composed of a prefusion-stabilized variant of the fusion (F) protein (DS-Cav1) and a CpG oligodeoxynucleotide adjuvant encapsulated within polyanhydride nanoparticles, termed RSVNanoVax. A prime-boost intranasal administration of RSVNanoVax in BALB/c mice significantly alleviated weight loss and pulmonary dysfunction in response to an RSV challenge, with protection maintained up to at least 6 mo postvaccination. In addition, vaccinated mice exhibited rapid viral clearance in the lungs as early as 2 d after RSV infection in both inbred and outbred populations. Vaccination induced tissue-resident memory CD4 and CD8 T cells in the lungs, as well as RSV F-directed neutralizing Abs. Based on the robust immune response elicited and the high level of durable protection observed, our prefusion RSV F nanovaccine is a promising new RSV vaccine candidate.
Collapse
Affiliation(s)
- Laura M Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
| | - Kathleen A Ross
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA.,Nanovaccine Institute, Ames, IA
| | - Kody A Waldstein
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
| | - Kevin L Legge
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA.,Nanovaccine Institute, Ames, IA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA.,Department of Pathology, University of Iowa, Iowa City, IA; and
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA.,Nanovaccine Institute, Ames, IA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA; .,Nanovaccine Institute, Ames, IA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA.,Department of Pathology, University of Iowa, Iowa City, IA; and
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Bhardwaj P, Bhatia E, Sharma S, Ahamad N, Banerjee R. Advancements in prophylactic and therapeutic nanovaccines. Acta Biomater 2020; 108:1-21. [PMID: 32268235 PMCID: PMC7163188 DOI: 10.1016/j.actbio.2020.03.020] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
Vaccines activate suitable immune responses to fight against diseases but can possess limitations such as compromised efficacy and immunogenic responses, poor stability, and requirement of adherence to multiple doses. ‘Nanovaccines’ have been explored to elicit a strong immune response with the advantages of nano-sized range, high antigen loading, enhanced immunogenicity, controlled antigen presentation, more retention in lymph nodes and promote patient compliance by a lower frequency of dosing. Various types of nanoparticles with diverse pathogenic or foreign antigens can help to overcome immunotolerance and alleviate the need of booster doses as required with conventional vaccines. Nanovaccines have the potential to induce both cell-mediated and antibody-mediated immunity and can render long-lasting immunogenic memory. With such properties, nanovaccines have shown high potential for the prevention of infectious diseases such as acquired immunodeficiency syndrome (AIDS), malaria, tuberculosis, influenza, and cancer. Their therapeutic potential has also been explored in the treatment of cancer. The various kinds of nanomaterials used for vaccine development and their effects on immune system activation have been discussed with special relevance to their implications in various pathological conditions. Statement of Significance Interaction of nanoparticles with the immune system has opened multiple avenues to combat a variety of infectious and non-infectious pathological conditions. Limitations of conventional vaccines have paved the path for nanomedicine associated benefits with a hope of producing effective nanovaccines. This review highlights the role of different types of nanovaccines and the role of nanoparticles in modulating the immune response of vaccines. The applications of nanovaccines in infectious and non-infectious diseases like malaria, tuberculosis, AIDS, influenza, and cancers have been discussed. It will help the readers develop an understanding of mechanisms of immune activation by nanovaccines and design appropriate strategies for novel nanovaccines.
Collapse
|
15
|
A single dose polyanhydride-based nanovaccine against paratuberculosis infection. NPJ Vaccines 2020; 5:15. [PMID: 32128256 PMCID: PMC7021715 DOI: 10.1038/s41541-020-0164-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 01/27/2020] [Indexed: 12/26/2022] Open
Abstract
Mycobacterium avium subsp. paratuberculosis (M. paratuberculosis) causes Johne’s disease in ruminants and is characterized by chronic gastroenteritis leading to heavy economic losses to the dairy industry worldwide. The currently available vaccine (inactivated bacterin in oil base) is not effective in preventing pathogen shedding and is rarely used to control Johne’s disease in dairy herds. To develop a better vaccine that can prevent the spread of Johne’s disease, we utilized polyanhydride nanoparticles (PAN) to encapsulate mycobacterial antigens composed of whole cell lysate (PAN-Lysate) and culture filtrate (PAN-Cf) of M. paratuberculosis. These nanoparticle-based vaccines (i.e., nanovaccines) were well tolerated in mice causing no inflammatory lesions at the site of injection. Immunological assays demonstrated a substantial increase in the levels of antigen-specific T cell responses post-vaccination in the PAN-Cf vaccinated group as indicated by high percentages of triple cytokine (IFN-γ, IL-2, TNF-α) producing CD8+ T cells. Following challenge, animals vaccinated with PAN-Cf continued to produce significant levels of double (IFN-γ, TNF-α) and single cytokine (IFN-γ) secreting CD8+ T cells compared with animals vaccinated with an inactivated vaccine. A significant reduction in bacterial load was observed in multiple organs of animals vaccinated with PAN-Cf, which is a clear indication of protection. Overall, the use of polyanhydride nanovaccines resulted in development of protective and sustained immunity against Johne’s disease, an approach that could be applied to counter other intracellular pathogens.
Collapse
|
16
|
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
|
17
|
Yan X, Zhou M, Yu S, Jin Z, Zhao K. An overview of biodegradable nanomaterials and applications in vaccines. Vaccine 2019; 38:1096-1104. [PMID: 31813649 DOI: 10.1016/j.vaccine.2019.11.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/30/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
Vaccination is the most cost-effective and sustainable way to prevent and eliminate infectious diseases. Compared with traditional vaccines, novel vaccines have better stability, longer duration and require less antigen usage. In addition, novel vaccines have better immune effects and significantly less toxic side effects. However, both novel vaccines and traditional vaccines require carrier molecules or adjuvants to produce an optimal immune response. There is an increasing demand for vaccine adjuvants and delivery systems that can induce stronger immune response whilst reducing production cost and the dose of vaccine. In recent years, nanotechnology has played an important role in the development of novel vaccine adjuvants and nano-delivery systems. Biodegradable materials have also received a lot of attention in medical science because they have excellent biocompatibility, biodegradability and low toxicity, which can protect antigens from degradation, increase antigen stability and provide slow release; resulting in enhanced immunogenicity. Therefore, biodegradable nanoparticles have attracted much attention in the formulation of vaccines. In this review, we outline some key features of biodegradable nanomaterials in the developing safer and more effective vaccines. The properties, structural characteristics, advantages and disadvantage of the biodegradable nanomaterials will be systematically reviewed. Additionally, applications, research progress and future prospects of biodegradable nanomaterials are discussed. This review will be help in future research work directed at developing biodegradable vaccine adjuvants or delivery carriers.
Collapse
Affiliation(s)
- Xingye Yan
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang University, Harbin 150080, China; Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin 150080, China
| | - Mo Zhou
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang University, Harbin 150080, China; Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin 150080, China
| | - Shuang Yu
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang University, Harbin 150080, China; Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin 150080, China
| | - Zheng Jin
- Key Laboratory of Chemical Engineering Process and Technology for High-efficiency Conversion, College of Chemistry and Material Sciences, Heilongjiang University, Harbin 150080, China
| | - Kai Zhao
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education, Heilongjiang University, Harbin 150080, China; Key Laboratory of Microbiology, College of Heilongjiang Province, School of Life Science, Heilongjiang University, Harbin 150080, China.
| |
Collapse
|
18
|
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
|
19
|
Ke X, Howard GP, Tang H, Cheng B, Saung MT, Santos JL, Mao HQ. Physical and chemical profiles of nanoparticles for lymphatic targeting. Adv Drug Deliv Rev 2019; 151-152:72-93. [PMID: 31626825 DOI: 10.1016/j.addr.2019.09.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/03/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022]
Abstract
Nanoparticles (NPs) have been gaining prominence as delivery vehicles for modulating immune responses to improve treatments against cancer and autoimmune diseases, enhancing tissue regeneration capacity, and potentiating vaccination efficacy. Various engineering approaches have been extensively explored to control the NP physical and chemical properties including particle size, shape, surface charge, hydrophobicity, rigidity and surface targeting ligands to modulate immune responses. This review examines a specific set of physical and chemical characteristics of NPs that enable efficient delivery targeted to secondary lymphoid tissues, specifically the lymph nodes and immune cells. A critical analysis of the structure-property-function relationship will facilitate further efforts to engineer new NPs with unique functionalities, identify novel utilities, and improve the clinical translation of NP formulations for immunotherapy.
Collapse
|
20
|
Prajapati SK, Jain A, Jain A, Jain S. Biodegradable polymers and constructs: A novel approach in drug delivery. Eur Polym J 2019; 120:109191. [DOI: 10.1016/j.eurpolymj.2019.08.018] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Kondakova OA, Nikitin NA, Evtushenko EA, Ryabchevskaya EM, Atabekov JG, Karpova OV. Vaccines against anthrax based on recombinant protective antigen: problems and solutions. Expert Rev Vaccines 2019; 18:813-828. [PMID: 31298973 DOI: 10.1080/14760584.2019.1643242] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Introduction: Anthrax is a dangerous bio-terror agent because Bacillus anthracis spores are highly resilient and can be easily aerosolized and disseminated. There is a threat of deliberate use of anthrax spores aerosol that could lead to serious fatal diseases outbreaks. Existing control measures against inhalation form of the disease are limited. All of this has provided an impetus to the development of new generation vaccines. Areas сovered: This review is devoted to challenges and achievements in the design of vaccines based on the anthrax recombinant protective antigen (rPA). Scientific databases have been searched, focusing on causes of PA instability and solutions to this problem, including new approaches of rPA expression, novel rPA-based vaccines formulations as well as the simultaneous usage of PA with other anthrax antigens. Expert opinion: PA is a central anthrax toxin component, playing a key role in the defense against encapsulated and unencapsulated strains. Subunit rPA-based vaccines have a good safety and protective profile. However, there are problems of PA instability that are greatly enhanced when using aluminum adjuvants. New adjuvant compositions, dry formulations and resistant to proteolysis and deamidation mutant PA forms can help to handle this issue. Devising a modern anthrax vaccine requires huge efforts.
Collapse
Affiliation(s)
- Olga A Kondakova
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Nikolai A Nikitin
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Ekaterina A Evtushenko
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Ekaterina M Ryabchevskaya
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Joseph G Atabekov
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Olga V Karpova
- a Department of Virology, Faculty of Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| |
Collapse
|
22
|
Al-Halifa S, Gauthier L, Arpin D, Bourgault S, Archambault D. Nanoparticle-Based Vaccines Against Respiratory Viruses. Front Immunol 2019; 10:22. [PMID: 30733717 PMCID: PMC6353795 DOI: 10.3389/fimmu.2019.00022] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/07/2019] [Indexed: 12/29/2022] Open
Abstract
The respiratory mucosa is the primary portal of entry for numerous viruses such as the respiratory syncytial virus, the influenza virus and the parainfluenza virus. These pathogens initially infect the upper respiratory tract and then reach the lower respiratory tract, leading to diseases. Vaccination is an affordable way to control the pathogenicity of viruses and constitutes the strategy of choice to fight against infections, including those leading to pulmonary diseases. Conventional vaccines based on live-attenuated pathogens present a risk of reversion to pathogenic virulence while inactivated pathogen vaccines often lead to a weak immune response. Subunit vaccines were developed to overcome these issues. However, these vaccines may suffer from a limited immunogenicity and, in most cases, the protection induced is only partial. A new generation of vaccines based on nanoparticles has shown great potential to address most of the limitations of conventional and subunit vaccines. This is due to recent advances in chemical and biological engineering, which allow the design of nanoparticles with a precise control over the size, shape, functionality and surface properties, leading to enhanced antigen presentation and strong immunogenicity. This short review provides an overview of the advantages associated with the use of nanoparticles as vaccine delivery platforms to immunize against respiratory viruses and highlights relevant examples demonstrating their potential as safe, effective and affordable vaccines.
Collapse
Affiliation(s)
- Soultan Al-Halifa
- Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC, Canada
| | - Laurie Gauthier
- Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC, Canada
- Département des Sciences Biologiques, Université du Québec à Montréal, Montreal, QC, Canada
- Faculté de Médecine Vétérinaire, Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC, Canada
| | - Dominic Arpin
- Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC, Canada
- Département des Sciences Biologiques, Université du Québec à Montréal, Montreal, QC, Canada
- Faculté de Médecine Vétérinaire, Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC, Canada
| | - Steve Bourgault
- Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC, Canada
- Faculté de Médecine Vétérinaire, Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC, Canada
| | - Denis Archambault
- Département des Sciences Biologiques, Université du Québec à Montréal, Montreal, QC, Canada
- Faculté de Médecine Vétérinaire, Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC, Canada
| |
Collapse
|
23
|
Karimi F, Alizadeh S, Alizadeh H. Immunogenicity of multi-walled carbon nanotubes functionalized with recombinant protective antigen domain 4 toward development of a nanovaccine against anthrax. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.07.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
24
|
Chen N, Gallovic MD, Tiet P, Ting JPY, Ainslie KM, Bachelder EM. Investigation of tunable acetalated dextran microparticle platform to optimize M2e-based influenza vaccine efficacy. J Control Release 2018; 289:114-124. [PMID: 30261204 DOI: 10.1016/j.jconrel.2018.09.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/08/2018] [Accepted: 09/22/2018] [Indexed: 01/26/2023]
Abstract
Influenza places a significant health and economic burden on society. Efficacy of seasonal influenza vaccines can be suboptimal due to poor matching between vaccine and circulating viral strains. An influenza vaccine that is broadly protective against multiple virus strains would significantly improve vaccine efficacy. The highly conserved ectodomain of matrix protein 2 (M2e) and 3'3' cyclic GMP-AMP (cGAMP) were selected as the antigen and adjuvant, respectively, to develop the basis for a potential universal influenza vaccine. The magnitude and kinetics of adaptive immune responses can have great impact on vaccine efficacy. M2e and cGAMP were therefore formulated within acetalated dextran (Ace-DEX) microparticles (MPs) of varying degradation profiles to examine the effect of differential vaccine delivery on humoral, cellular, and protective immunity. All Ace-DEX MP vaccines containing M2e and cGAMP elicited potent humoral and cellular responses in vivo and offered substantial protection against a lethal influenza challenge, suggesting significant vaccine efficacy. Serum antibodies from Ace-DEX MP vaccinated mice also demonstrated cross reactivity against M2e sequences of various viral strains, which indicates the potential for broadly protective immunity. Of all the formulations tested, the slowest-degrading M2e or cGAMP MPs elicited the greatest antibody production, cellular response, and protection against a viral challenge. This indicated the importance of flexible control over antigen and adjuvant delivery. Overall, robust immune responses, cross reactivity against multiple viral strains, and tunable delivery profiles make the Ace-DEX MP platform a powerful subunit vaccine delivery system.
Collapse
Affiliation(s)
- Naihan Chen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Matthew D Gallovic
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Pamela Tiet
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Jenny P-Y Ting
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA; Institute for Inflammatory Diseases, University of North Carolina, Chapel Hill, NC, USA; Center for Translational Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
25
|
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
|
26
|
Wagner-Muñiz DA, Haughney SL, Kelly SM, Wannemuehler MJ, Narasimhan B. Room Temperature Stable PspA-Based Nanovaccine Induces Protective Immunity. Front Immunol 2018; 9:325. [PMID: 29599766 PMCID: PMC5863507 DOI: 10.3389/fimmu.2018.00325] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/06/2018] [Indexed: 01/05/2023] Open
Abstract
Streptococcus pneumoniae is a major causative agent of pneumonia, a debilitating disease particularly in young and elderly populations, and is the leading worldwide cause of death in children under the age of five. While there are existing vaccines against S. pneumoniae, none are protective across all serotypes. Pneumococcal surface protein A (PspA), a key virulence factor of S. pneumoniae, is an antigen that may be incorporated into future vaccines to address the immunological challenges presented by the diversity of capsular antigens. PspA has been shown to be immunogenic and capable of initiating a humoral immune response that is reactive across approximately 94% of pneumococcal strains. Biodegradable polyanhydrides have been studied as a nanoparticle-based vaccine (i.e., nanovaccine) platform to stabilize labile proteins, to provide adjuvanticity, and enhance patient compliance by providing protective immunity in a single dose. In this study, we designed a room temperature stable PspA-based polyanhydride nanovaccine that eliminated the need for a free protein component (i.e., 100% encapsulated within the nanoparticles). Mice were immunized once with the lead nanovaccine and upon challenge, presented significantly higher survival rates than animals immunized with soluble protein alone, even with a 25-fold reduction in protein dose. This lead nanovaccine formulation performed similarly to protein adjuvanted with Alum, however, with much less tissue reactogenicity at the site of immunization. By eliminating the free PspA from the nanovaccine formulation, the lead nanovaccine was efficacious after being stored dry for 60 days at room temperature, breaking the need for maintaining the cold chain. Altogether, this study demonstrated that a single dose PspA-based nanovaccine against S. pneumoniae induced protective immunity and provided thermal stability when stored at room temperature for at least 60 days.
Collapse
Affiliation(s)
- Danielle A. Wagner-Muñiz
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Shannon L. Haughney
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Sean M. Kelly
- Department of Chemical and Biological Engineering, 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
|
27
|
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: 49] [Impact Index Per Article: 7.0] [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
|
28
|
Phanse Y, Carrillo-Conde BR, Ramer-Tait AE, Roychoudhury R, Broderick S, Pohl N, Rajan K, Narasimhan B, Wannemuehler MJ, Bellaire BH. Functionalization promotes pathogen-mimicking characteristics of polyanhydride nanoparticle adjuvants. J Biomed Mater Res A 2017; 105:2762-2771. [PMID: 28556563 DOI: 10.1002/jbm.a.36128] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/08/2017] [Accepted: 05/23/2017] [Indexed: 11/08/2022]
Abstract
Rational design of adjuvants and delivery systems will promote development of next-generation vaccines to control emerging and re-emerging diseases. To accomplish this, understanding the immune-enhancing properties of new adjuvants relative to those induced by natural infections can help with the development of pathogen-mimicking materials that will effectively initiate innate immune signaling cascades. In this work, the surfaces of polyanhydride nanoparticles composed of sebacic acid (SA) and 1,6-bis(p-carboxyphenoxy) hexane were decorated with an ethylene diamine spacer partially modified with either a glycolic acid linker or an α-1,2-linked di-mannopyranoside (di-mannose) to confer "pathogen-like" properties and enhance adjuvanticity. Co-incubation of linker-modified nanoparticles with dendritic cells (DCs) elicited significant increases in surface expression of MHC I, MHC II, CD86, and CD40, and enhanced secretion of IL-6, IL-12p40, and TNF-α. An 800% increase in uptake of ethylene-diamine-spaced, linker and di-mannose functionalized polyanhydride nanoparticles was also observed. Together, our data showed that linker-functionalized polyanhydride nanoparticles demonstrate similar patterns of uptake, intracellular trafficking, particle persistence, and innate activation as did DCs exposed to Yersinia pestis or Escherichia coli. These results set the stage for rational selection of adjuvant chemistries to induce pathogen-mimicking immune responses. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2762-2771, 2017.
Collapse
Affiliation(s)
- Yashdeep Phanse
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Wisconsin-Madison, Wisconsin, 53706
| | | | - Amanda E Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Nebraska, 68588
| | - Rajarshi Roychoudhury
- Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana, 47401
| | - Scott Broderick
- Department of Materials Design and Innovation, University at Buffalo, Buffalo, 14260, New York
| | - Nicola Pohl
- Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana, 47401
| | - Krishna Rajan
- Department of Materials Design and Innovation, University at Buffalo, Buffalo, 14260, New York
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa
| | - Bryan H Bellaire
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa
| |
Collapse
|
29
|
Gallovic MD, Schully KL, Bell MG, Elberson MA, Palmer JR, Darko CA, Bachelder EM, Wyslouzil BE, Keane-Myers AM, Ainslie KM. Acetalated Dextran Microparticulate Vaccine Formulated via Coaxial Electrospray Preserves Toxin Neutralization and Enhances Murine Survival Following Inhalational Bacillus Anthracis Exposure. Adv Healthc Mater 2016; 5:2617-2627. [PMID: 27594343 DOI: 10.1002/adhm.201600642] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 07/20/2016] [Indexed: 12/30/2022]
Abstract
Subunit formulations are regarded as the safest type of vaccine, but they often contain a protein-based antigen that can result in significant challenges, such as preserving antigenicity during formulation and administration. Many studies have demonstrated that encapsulation of protein antigens in polymeric microparticles (MPs) via emulsion techniques results in total IgG antibody titers comparable to alum formulations, however, the antibodies themselves are non-neutralizing. To address this issue, a coaxial electrohydrodynamic spraying (electrospray) technique is used to formulate a microparticulate-based subunit anthrax vaccine under conditions that minimize recombinant protective antigen (rPA) exposure to harsh solvents and high shear stress. rPA and the adjuvant resiquimod are encapsulated either in separate or the same acetalated dextran MPs. Using a murine model, the electrospray formulations lead to higher IgG2a subtype titers as well as comparable total IgG antibody titers and toxin neutralization relative to the FDA-approved vaccine (BioThrax). BioThrax provides no protection against a lethal inhalational challenge of the highly virulent Ames Bacillus anthracis anthrax strain, whereas 50% of the mice vaccinated with separately encapsulated electrospray MPs survive. Overall, this study demonstrates the potential use of electrospray for encapsulating protein antigens in polymeric MPs.
Collapse
Affiliation(s)
- Matthew D. Gallovic
- Department of Chemical and Biomolecular Engineering; College of Engineering; The Ohio State University; Columbus OH 43210 USA
- Division of Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina; Chapel Hill NC 27599 USA
| | - Kevin L. Schully
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Matthew G. Bell
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Margaret A. Elberson
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - John R. Palmer
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Christian A. Darko
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Eric M. Bachelder
- Division of Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina; Chapel Hill NC 27599 USA
| | - Barbara E. Wyslouzil
- Department of Chemical and Biomolecular Engineering; College of Engineering; The Ohio State University; Columbus OH 43210 USA
- Department of Chemistry and Biochemistry; College of Arts and Sciences; The Ohio State University; Columbus OH 43210 USA
| | - Andrea M. Keane-Myers
- Vaccine and Medical Countermeasures Department; Biological Defense Research Directorate; Naval Medical Research Center; Fort Detrick MD 20910 USA
| | - Kristy M. Ainslie
- Division of Molecular Pharmaceutics; Eshelman School of Pharmacy; University of North Carolina; Chapel Hill NC 27599 USA
| |
Collapse
|
30
|
Abstract
This review focusses on polyanhydrides, a fascinating class of degradable polymers that have been used in and investigated for many bio-related applications because of their degradability and capacity to undergo surface erosion. This latter phenomenon is driven by hydrolysis of the anhydride moieties at the surface and high hydrophobicity of the polymer such that degradation and mass loss (erosion) occur before water can penetrate deep within the bulk of the polymer. As such, when surface-eroding polymers are used as therapeutic delivery vehicles, the rate of delivery is often controlled by the rate of polymer erosion, providing predictable and controlled release rates that are often zero-order. These desirable attributes are heavily influenced by polymer composition and morphology, and therefore also monomer structure and polymerization method. This review examines approaches for polyanhydride synthesis, discusses their general thermomechanical properties, surveys their hydrolysis and degradation processes along with their biocompatibility, and looks at recent developments and uses of polyanhydrides in drug delivery, stimuli-responsive materials, and novel nanotechnologies.
Collapse
|
31
|
Renukaradhya GJ, Narasimhan B, Mallapragada SK. Respiratory nanoparticle-based vaccines and challenges associated with animal models and translation. J Control Release 2015; 219:622-631. [PMID: 26410807 PMCID: PMC4760633 DOI: 10.1016/j.jconrel.2015.09.047] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 12/14/2022]
Abstract
Vaccine development has had a huge impact on human health. However, there is a significant need to develop efficacious vaccines for several existing as well as emerging respiratory infectious diseases. Several challenges need to be overcome to develop efficacious vaccines with translational potential. This review focuses on two aspects to overcome some barriers - 1) the development of nanoparticle-based vaccines, and 2) the choice of suitable animal models for respiratory infectious diseases that will allow for translation. Nanoparticle-based vaccines, including subunit vaccines involving synthetic and/or natural polymeric adjuvants and carriers, as well as those based on virus-like particles offer several key advantages to help overcome the barriers to effective vaccine development. These include the ability to deliver combinations of antigens, target the vaccine formulation to specific immune cells, enable cross-protection against divergent strains, act as adjuvants or immunomodulators, allow for sustained release of antigen, enable single dose delivery, and potentially obviate the cold chain. While mouse models have provided several important insights into the mechanisms of infectious diseases, they are often a limiting step in translation of new vaccines to the clinic. An overview of different animal models involved in vaccine research for respiratory infections, with advantages and disadvantages of each model, is discussed. Taken together, advances in nanotechnology, combined with the right animal models for evaluating vaccine efficacy, has the potential to revolutionize vaccine development for respiratory infections.
Collapse
Affiliation(s)
- Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, United States
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
32
|
Binnebose AM, Haughney SL, Martin R, Imerman PM, Narasimhan B, Bellaire BH. Polyanhydride Nanoparticle Delivery Platform Dramatically Enhances Killing of Filarial Worms. PLoS Negl Trop Dis 2015; 9:e0004173. [PMID: 26496201 PMCID: PMC4619673 DOI: 10.1371/journal.pntd.0004173] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 09/28/2015] [Indexed: 11/18/2022] Open
Abstract
Filarial diseases represent a significant social and economic burden to over 120 million people worldwide and are caused by endoparasites that require the presence of symbiotic bacteria of the genus Wolbachia for fertility and viability of the host parasite. Targeting Wolbachia for elimination is a therapeutic approach that shows promise in the treatment of onchocerciasis and lymphatic filariasis. Here we demonstrate the use of a biodegradable polyanhydride nanoparticle-based platform for the co-delivery of the antibiotic doxycycline with the antiparasitic drug, ivermectin, to reduce microfilarial burden and rapidly kill adult worms. When doxycycline and ivermectin were co-delivered within polyanhydride nanoparticles, effective killing of adult female Brugia malayi filarial worms was achieved with approximately 4,000-fold reduction in the amount of drug used. Additionally the time to death of the macrofilaria was also significantly reduced (five-fold) when the anti-filarial drug cocktail was delivered within polyanhydride nanoparticles. We hypothesize that the mechanism behind this dramatically enhanced killing of the macrofilaria is the ability of the polyanhydride nanoparticles to behave as a Trojan horse and penetrate the cuticle, bypassing excretory pumps of B. malayi, and effectively deliver drug directly to both the worm and Wolbachia at high enough microenvironmental concentrations to cause death. These provocative findings may have significant consequences for the reduction in the amount of drug and the length of treatment required for filarial infections in terms of patient compliance and reduced cost of treatment. Infection with the filarial endoparasites Brugia malayi and its symbiotic bacteria Wolbachia represent a significant burden to both humans and animals. Current treatment protocols include use of multiple drugs over a course of months to years, resulting in high costs, undesirable side effects, and poor patient compliance. By encapsulating two of these drugs, ivermectin and doxycycline, into biodegradable polyanhydride nanoparticles, we report the ability to effectively kill adult B. malayi with up to a 4,000-fold reduction in the amount of drug used. These results demonstrate a promising role for the use of nanoscale drug carriers to reduce both the course of treatment and the amount of drug needed to increase affordability of lymphatic filariasis treatment and enhance patient compliance.
Collapse
Affiliation(s)
- Andrea M. Binnebose
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa, United States of America
| | - Shannon L. Haughney
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Richard Martin
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Paula M. Imerman
- Veterinary Diagnostic Laboratory, Iowa State University, Ames, Iowa, United States of America
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, United States of America
- * E-mail: (BN); (BHB)
| | - Bryan H. Bellaire
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa, United States of America
- * E-mail: (BN); (BHB)
| |
Collapse
|
33
|
McHugh KJ, Guarecuco R, Langer R, Jaklenec A. Single-injection vaccines: Progress, challenges, and opportunities. J Control Release 2015; 219:596-609. [PMID: 26254198 DOI: 10.1016/j.jconrel.2015.07.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/27/2015] [Accepted: 07/28/2015] [Indexed: 01/01/2023]
Abstract
Currently, vaccination is the most efficient and cost-effective medical treatment for infectious diseases; however, each year 10 million infants remain underimmunized due to current vaccination schedules that require multiple doses to be administered across months or years. These dosing regimens are especially challenging in the developing world where limited healthcare access poses a major logistical barrier to immunization. Over the past four decades, researchers have attempted to overcome this issue by developing single-administration vaccines based on controlled-release antigen delivery systems. These systems can be administered once, but release antigen over an extended period of time to elicit both primary and secondary immune responses resulting in antigen-specific immunological memory. Unfortunately, unlike controlled release systems for drugs, single-administration vaccines have yet to be commercialized due to poor antigen stability and difficulty in obtaining unconventional release kinetics. This review discusses the current state of single-administration vaccination, challenges delaying the development of these vaccines, and potential strategies for overcoming these challenges.
Collapse
Affiliation(s)
- Kevin J McHugh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Rohiverth Guarecuco
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States.
| |
Collapse
|
34
|
Poetz KL, Mohammed HS, Shipp DA. Surface Eroding, Semicrystalline Polyanhydrides via Thiol–Ene “Click” Photopolymerization. Biomacromolecules 2015; 16:1650-9. [DOI: 10.1021/acs.biomac.5b00280] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Katie L. Poetz
- Department
of Chemistry and Biomolecular Science and ‡Center for Advanced Materials Processing, Clarkson University, Potsdam, New York 13699-5810, United States
| | - Halimatu S. Mohammed
- Department
of Chemistry and Biomolecular Science and ‡Center for Advanced Materials Processing, Clarkson University, Potsdam, New York 13699-5810, United States
| | - Devon A. Shipp
- Department
of Chemistry and Biomolecular Science and ‡Center for Advanced Materials Processing, Clarkson University, Potsdam, New York 13699-5810, United States
| |
Collapse
|
35
|
Sustained release and stabilization of therapeutic antibodies using amphiphilic polyanhydride nanoparticles. Chem Eng Sci 2015. [DOI: 10.1016/j.ces.2014.08.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Ross KA, Loyd H, Wu W, Huntimer L, Ahmed S, Sambol A, Broderick S, Flickinger Z, Rajan K, Bronich T, Mallapragada S, Wannemuehler MJ, Carpenter S, Narasimhan B. Hemagglutinin-based polyanhydride nanovaccines against H5N1 influenza elicit protective virus neutralizing titers and cell-mediated immunity. Int J Nanomedicine 2014; 10:229-43. [PMID: 25565816 PMCID: PMC4284014 DOI: 10.2147/ijn.s72264] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
H5N1 avian influenza is a significant global concern with the potential to become the next pandemic threat. Recombinant subunit vaccines are an attractive alternative for pandemic vaccines compared to traditional vaccine technologies. In particular, polyanhydride nanoparticles encapsulating subunit proteins have been shown to enhance humoral and cell-mediated immunity and provide protection upon lethal challenge. In this work, a recombinant H5 hemagglutinin trimer (H5₃) was produced and encapsulated into polyanhydride nanoparticles. The studies performed indicated that the recombinant H5₃ antigen was a robust immunogen. Immunizing mice with H5₃ encapsulated into polyanhydride nanoparticles induced high neutralizing antibody titers and enhanced CD4(+) T cell recall responses in mice. Finally, the H5₃-based polyanhydride nanovaccine induced protective immunity against a low-pathogenic H5N1 viral challenge. Informatics analyses indicated that mice receiving the nanovaccine formulations and subsequently challenged with virus were similar to naïve mice that were not challenged. The current studies provide a basis to further exploit the advantages of polyanhydride nanovaccines in pandemic scenarios.
Collapse
Affiliation(s)
- Kathleen A Ross
- Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Hyelee Loyd
- Animal Science, Iowa State University, Ames, IA, USA
| | - Wuwei Wu
- Animal Science, Iowa State University, Ames, IA, USA
| | - Lucas Huntimer
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, USA
| | - Shaheen Ahmed
- Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anthony Sambol
- Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Scott Broderick
- Materials Science and Engineering, Iowa State University, Ames, IA, USA
| | | | - Krishna Rajan
- Materials Science and Engineering, Iowa State University, Ames, IA, USA
| | - Tatiana Bronich
- Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surya Mallapragada
- Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | | | | | - Balaji Narasimhan
- Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| |
Collapse
|
37
|
Sharma R, Agrawal U, Mody N, Vyas SP. Polymer nanotechnology based approaches in mucosal vaccine delivery: challenges and opportunities. Biotechnol Adv 2014; 33:64-79. [PMID: 25499178 DOI: 10.1016/j.biotechadv.2014.12.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 11/20/2014] [Accepted: 12/05/2014] [Indexed: 01/11/2023]
Abstract
Mucosal sites serve as the main portal for the entry of pathogens and thus immunization through mucosal routes can greatly improve the immunity. Researchers are continuously exploring the vaccination strategies to engender protective mucosal immune responses. Unearthing of mucosal adjuvants, that are safe and effective, is enhancing the magnitude and quality of the protective immune response. Use of nanotechnology based polymeric nanocarrier systems which encapsulate vaccine components for protection of sensitive payload, incorporate mucosal adjuvants to maximize the immune responses and target the mucosal immune system is a key strategy to improve the effectiveness of mucosal vaccines. These advances promise to accelerate the development and testing of new mucosal vaccines against many human diseases. This review focuses on the need for the development of nanocarrier based mucosal vaccines with emphases on the polymeric nanoparticles, their clinical status and future perspectives. This review focuses on the need and new insights for the development of nanoarchitecture governed mucosal vaccination with emphases on the various polymeric nanoparticles, their clinical status and future perspectives.
Collapse
Affiliation(s)
- Rajeev Sharma
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Vishwavidyalaya, Sagar, M.P. 470003 India.
| | - Udita Agrawal
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Vishwavidyalaya, Sagar, M.P. 470003 India.
| | - Nishi Mody
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Vishwavidyalaya, Sagar, M.P. 470003 India.
| | - Suresh P Vyas
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Vishwavidyalaya, Sagar, M.P. 470003 India.
| |
Collapse
|
38
|
Vela-Ramirez JE, Goodman JT, Boggiatto PM, Roychoudhury R, Pohl NLB, Hostetter JM, Wannemuehler MJ, Narasimhan B. Safety and biocompatibility of carbohydrate-functionalized polyanhydride nanoparticles. AAPS JOURNAL 2014; 17:256-67. [PMID: 25421457 DOI: 10.1208/s12248-014-9699-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/07/2014] [Indexed: 01/08/2023]
Abstract
Carbohydrate functionalization of nanoparticles allows for targeting of C-type lectin receptors. This family of pattern recognition receptors expressed on innate immune cells, such as macrophages and dendritic cells, can be used to modulate immune responses. In this work, the in vivo safety profile of carbohydrate-functionalized polyanhydride nanoparticles was analyzed following parenteral and intranasal administration in mice. Polyanhydride nanoparticles based on 1,6-bis-(p-carboxyphenoxy)hexane and 1,8-bis-(p-carboxyphenoxy)-3,6-dioxaoctane were used. Nanoparticle functionalization with di-mannose (specifically carboxymethyl-α-D-mannopyranosyl-(1,2)-D-mannopyranoside), galactose (specifically carboxymethyl-β-galactoside), or glycolic acid induced no adverse effects after administration based on histopathological evaluation of liver, kidneys, and lungs. Regardless of the polymer formulation, there was no evidence of hepatic or renal damage or dysfunction observed in serum or urine samples. The histological profile of cellular infiltration and the cellular distribution and kinetics in the lungs of mice administered with nanoparticle treatments followed similar behavior as that observed in the lungs of animals administered with saline. Cytokine and chemokine profiles in bronchoalveolar lavage fluid indicated surface chemistry dependence on modest secretion of IL-6, IP-10, and MCP-1; however, there was no evidence of any deleterious histopathological changes. Based on these analyses, carbohydrate-functionalized nanoparticles are safe for in vivo applications. These results provide foundational information towards the evaluation of the capabilities of these surface-modified nanoparticles as vaccine delivery formulations.
Collapse
Affiliation(s)
- Julia E Vela-Ramirez
- Department of Chemical and Biological Engineering, Iowa State University, 2035 Sweeney Hall, Ames, Iowa, 50011, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Haughney SL, Ross KA, Boggiatto PM, Wannemuehler MJ, Narasimhan B. Effect of nanovaccine chemistry on humoral immune response kinetics and maturation. NANOSCALE 2014; 6:13770-13778. [PMID: 25285425 DOI: 10.1039/c4nr03724c] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Acute respiratory infections represent a significant portion of global morbidity and mortality annually. There is a critical need for efficacious vaccines against respiratory pathogens. To vaccinate against respiratory disease, pulmonary delivery is an attractive route because it mimics the route of natural infection and can confer both mucosal and systemic immunity. We have previously demonstrated that a single dose, intranasal vaccine based on polyanhydride nanoparticles elicited a protective immune response against Yersinia pestis for at least 40 weeks after immunization with F1-V. Herein, we investigate the effect of nanoparticle chemistry and its attributes on the kinetics and maturation of the antigen-specific serum antibody response. We demonstrate that manipulation of polyanhydride nanoparticle chemistry facilitated differential kinetics of development of antibody titers, avidity, and epitope specificity. The results provide new insights into the underlying role(s) of nanoparticle chemistry in providing long-lived humoral immunity and aid in the rational design of nanovaccine formulations to induce long-lasting and mature antibody responses.
Collapse
Affiliation(s)
- Shannon L Haughney
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA.
| | | | | | | | | |
Collapse
|
40
|
Vela Ramirez JE, Roychoudhury R, Habte HH, Cho MW, Pohl NLB, Narasimhan B. Carbohydrate-functionalized nanovaccines preserve HIV-1 antigen stability and activate antigen presenting cells. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2014; 25:1387-406. [PMID: 25068589 DOI: 10.1080/09205063.2014.940243] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The functionalization of polymeric nanoparticles with ligands that target specific receptors on immune cells offers the opportunity to tailor adjuvant properties by conferring pathogen mimicking attributes to the particles. Polyanhydride nanoparticles are promising vaccine adjuvants with desirable characteristics such as immunomodulation, sustained antigen release, activation of antigen presenting cells (APCs), and stabilization of protein antigens. These capabilities can be exploited to design nanovaccines against viral pathogens, such as HIV-1, due to the important role of dendritic cells (DCs) and macrophages in viral spread. In this work, an optimized process was developed for carbohydrate functionalization of HIV-1 antigen-loaded polyanhydride nanoparticles. The carbohydrate-functionalized nanoparticles preserved antigenic properties upon release and also enabled sustained antigen release kinetics. Particle internalization was observed to be chemistry-dependent with positively charged nanoparticles being taken up more efficiently by DCs. Up-regulation of the activation makers CD40 and CD206 was demonstrated with carboxymethyl-α-d-mannopyranosyl-(1,2)-d-mannopyranoside functionalized nanoparticles. The secretion of the cytokines IL-6 and TNF-α was shown to be chemistry-dependent upon stimulation with carbohydrate-functionalized nanoparticles. These results offer important new insights upon the interactions between carbohydrate-functionalized nanoparticles and APCs and provide foundational information for the rational design of targeted nanovaccines against HIV-1.
Collapse
Affiliation(s)
- J E Vela Ramirez
- a Department of Chemical and Biological Engineering , Iowa State University , Ames , IA 50011 , USA
| | | | | | | | | | | |
Collapse
|
41
|
Adamo R. Glycan surface antigens fromBacillus anthracisas vaccine targets: current status and future perspectives. Expert Rev Vaccines 2014; 13:895-907. [DOI: 10.1586/14760584.2014.924404] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
42
|
Kaur M, Singh S, Bhatnagar R. Anthrax vaccines: present status and future prospects. Expert Rev Vaccines 2014; 12:955-70. [PMID: 23984963 DOI: 10.1586/14760584.2013.814860] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The management of anthrax remains a top priority among the biowarfare/bioterror agents. It was the Bacillus anthracis spore attack through the US mail system after the September 11, 2001, terrorist attacks in the USA that highlighted the potential of B. anthracis as a bioterrorism agent and the threat posed by its deliberate dissemination. These attacks invigorated the efforts toward understanding the anthrax pathogenesis and development of more comprehensive medical intervention strategies for its containment in case of both natural disease and manmade, accidental or deliberate infection of a non-suspecting population. Currently, efforts are directed toward the development of safe and efficacious vaccines as well as intervention tools for controlling the disease in the advanced fulminant stage when toxemia has already developed. This work presents an overview of the current understanding of anthrax pathogenesis and recent advances made, particularly after 2001, for the successful management of anthrax and outlines future perspectives.
Collapse
Affiliation(s)
- Manpreet Kaur
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, Delhi, India
| | | | | |
Collapse
|
43
|
Ross KA, Loyd H, Wu W, Huntimer L, Wannemuehler MJ, Carpenter S, Narasimhan B. Structural and antigenic stability of H5N1 hemagglutinin trimer upon release from polyanhydride nanoparticles. J Biomed Mater Res A 2014; 102:4161-8. [DOI: 10.1002/jbm.a.35086] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 12/19/2013] [Accepted: 01/15/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Kathleen A. Ross
- Department of Chemical and Biological Engineering; Iowa State University; Ames Iowa 50011
| | - Hyelee Loyd
- Department of Animal Science; Iowa State University; Ames Iowa 50011
| | - Wuwei Wu
- Department of Animal Science; Iowa State University; Ames Iowa 50011
| | - Lucas Huntimer
- Department of Veterinary Microbiology and Preventive Medicine; Iowa State University; Ames Iowa 50011
| | - Michael J. Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine; Iowa State University; Ames Iowa 50011
| | - Susan Carpenter
- Department of Animal Science; Iowa State University; Ames Iowa 50011
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering; Iowa State University; Ames Iowa 50011
| |
Collapse
|
44
|
Phanse Y, Carrillo-Conde BR, Ramer-Tait AE, Broderick S, Kong CS, Rajan K, Flick R, Mandell RB, Narasimhan B, Wannemuehler MJ. A systems approach to designing next generation vaccines: combining α-galactose modified antigens with nanoparticle platforms. Sci Rep 2014; 4:3775. [PMID: 24441019 PMCID: PMC3895907 DOI: 10.1038/srep03775] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/20/2013] [Indexed: 12/21/2022] Open
Abstract
Innovative vaccine platforms are needed to develop effective countermeasures against emerging and re-emerging diseases. These platforms should direct antigen internalization by antigen presenting cells and promote immunogenic responses. This work describes an innovative systems approach combining two novel platforms, αGalactose (αGal)-modification of antigens and amphiphilic polyanhydride nanoparticles as vaccine delivery vehicles, to rationally design vaccine formulations. Regimens comprising soluble αGal-modified antigen and nanoparticle-encapsulated unmodified antigen induced a high titer, high avidity antibody response with broader epitope recognition of antigenic peptides than other regimen. Proliferation of antigen-specific CD4+ T cells was also enhanced compared to a traditional adjuvant. Combining the technology platforms and augmenting immune response studies with peptide arrays and informatics analysis provides a new paradigm for rational, systems-based design of next generation vaccine platforms against emerging and re-emerging pathogens.
Collapse
Affiliation(s)
- Yashdeep Phanse
- 1] Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011 [2]
| | - Brenda R Carrillo-Conde
- 1] Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011 [2]
| | - Amanda E Ramer-Tait
- 1] Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011 [2] Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68583
| | - Scott Broderick
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Chang Sun Kong
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Krishna Rajan
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011
| | - Ramon Flick
- BioProtection Systems Corporation, a subsidiary of NewLink Genetics Corporation, Ames, IA 50010
| | - Robert B Mandell
- 1] BioProtection Systems Corporation, a subsidiary of NewLink Genetics Corporation, Ames, IA 50010 [2]
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011
| |
Collapse
|
45
|
|
46
|
Multifunctional nanoparticles for targeted delivery of immune activating and cancer therapeutic agents. J Control Release 2013; 172:1020-34. [PMID: 24140748 DOI: 10.1016/j.jconrel.2013.10.012] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/08/2013] [Accepted: 10/09/2013] [Indexed: 01/20/2023]
Abstract
Nanoparticles (NPs) have been extensively investigated for applications in both experimental and clinical settings to improve delivery efficiency of therapeutic and diagnostic agents. Most recently, novel multifunctional nanoparticles have attracted much attention because of their ability to carry diverse functionalities to achieve effective synergistic therapeutic treatments. Multifunctional NPs have been designed to co-deliver multiple components, target the delivery of drugs by surface functionalization, and realize therapy and diagnosis simultaneously. In this review, various materials of diverse chemistries for fabricating multifunctional NPs with distinctive architectures are discussed and compared. Recent progress involving multifunctional NPs for immune activation, anticancer drug delivery, and synergistic theranostics is the focus of this review. Overall, this comprehensive review demonstrates that multifunctional NPs have distinctive properties that make them highly suitable for targeted therapeutic delivery in these areas.
Collapse
|
47
|
Haughney SL, Petersen LK, Schoofs AD, Ramer-Tait AE, King JD, Briles DE, Wannemuehler MJ, Narasimhan B. Retention of structure, antigenicity, and biological function of pneumococcal surface protein A (PspA) released from polyanhydride nanoparticles. Acta Biomater 2013; 9:8262-71. [PMID: 23774257 DOI: 10.1016/j.actbio.2013.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/29/2013] [Accepted: 06/03/2013] [Indexed: 02/03/2023]
Abstract
Pneumococcal surface protein A (PspA) is a choline-binding protein which is a virulence factor found on the surface of all Streptococcus pneumoniae strains. Vaccination with PspA has been shown to be protective against a lethal challenge with S. pneumoniae, making it a promising immunogen for use in vaccines. Herein the design of a PspA-based subunit vaccine using polyanhydride nanoparticles as a delivery platform is described. Nanoparticles based on sebacic acid (SA), 1,6-bis-(p-carboxyphenoxy)hexane (CPH) and 1,8-bis-(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG), specifically 50:50 CPTEG:CPH and 20:80 CPH:SA, were used to encapsulate and release PspA. The protein released from the nanoparticle formulations retained its primary and secondary structure as well as its antigenicity. The released PspA was also biologically functional based on its ability to bind to apolactoferrin and prevent its bactericidal activity against Escherichia coli. When the PspA nanoparticle formulations were administered subcutaneously to mice they elicited a high titer and high avidity anti-PspA antibody response. Together these studies provide a framework for the rational design of a vaccine against S. pneumoniae based on polyanhydride nanoparticles.
Collapse
|
48
|
Petersen LK, Huntimer L, Walz K, Ramer-Tait A, Wannemuehler MJ, Narasimhan B. Combinatorial evaluation of in vivo distribution of polyanhydride particle-based platforms for vaccine delivery. Int J Nanomedicine 2013; 8:2213-25. [PMID: 23818778 PMCID: PMC3693819 DOI: 10.2147/ijn.s45317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Several challenges are associated with current vaccine strategies, including repeated immunizations, poor patient compliance, and limited approved routes for delivery, which may hinder induction of protective immunity. Thus, there is a need for new vaccine adjuvants capable of multi-route administration and prolonged antigen release at the site of administration by providing a depot within tissue. In this work, we designed a combinatorial platform to investigate the in vivo distribution, depot effect, and localized persistence of polyanhydride nanoparticles as a function of nanoparticle chemistry and administration route. Our observations indicated that the route of administration differentially affected tissue residence times. All nanoparticles rapidly dispersed when delivered intranasally but provided a depot when administered parenterally. When amphiphilic and hydrophobic nanoparticles were administered intranasally, they persisted within lung tissue. These results provide insights into the chemistry- and route-dependent distribution and tissue-specific association of polyanhydride nanoparticle-based vaccine adjuvants.
Collapse
Affiliation(s)
- Latrisha K Petersen
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | | | |
Collapse
|
49
|
Joshi VB, Geary SM, Carrillo-Conde BR, Narasimhan B, Salem AK. Characterizing the antitumor response in mice treated with antigen-loaded polyanhydride microparticles. Acta Biomater 2013; 9:5583-9. [PMID: 23153760 PMCID: PMC3562412 DOI: 10.1016/j.actbio.2012.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 10/19/2012] [Accepted: 11/01/2012] [Indexed: 12/31/2022]
Abstract
Delivery of vaccine antigens with an appropriate adjuvant can trigger potential immune responses against cancer leading to reduced tumor growth and improved survival. In this study, various formulations of a bioerodible amphiphilic polyanhydride copolymer based on 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG) and 1,6-bis(p-carboxyphenoxy) hexane (CPH) with inherent adjuvant properties were evaluated for antigen-loading properties, immunogenicity and antitumor activity. Mice were vaccinated with 50:50 CPTEG:CPH microparticles encapsulating a model tumor antigen, ovalbumin (OVA), in combination with the Toll-like receptor-9 agonist, CpG oligonucleotide 1826 (CpG ODN). Mice treated with OVA-encapsulated CPTEG:CPH particles elicited the highest CD8(+) T cell responses on days 14 and 20 when compared to other treatment groups. This treatment group also displayed the most delayed tumor progression and the most extended survival times. Particles encapsulating OVA and CpG ODN generated the highest anti-OVA IgG(1) antibody responses in mice but these mice did not show significant tumor protection. These results suggest that antigen-loaded CPTEG:CPH microparticles can stimulate antigen-specific cellular responses and could therefore potentially be used to promote antitumor responses in cancer patients.
Collapse
Affiliation(s)
- Vijaya B. Joshi
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242
| | - Sean M. Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242
| | | | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011
| | - Aliasger K. Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242
| |
Collapse
|