1
|
Euliano EM, Pogostin BH, Agrawal A, Yu MH, Baryakova TH, Graf TP, Kunkel AA, Cahue KA, Hartgerink JD, McHugh KJ. A TLR7 Agonist Conjugated to a Nanofibrous Peptide Hydrogel as a Potent Vaccine Adjuvant. Adv Healthc Mater 2024:e2402958. [PMID: 39460390 DOI: 10.1002/adhm.202402958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Indexed: 10/28/2024]
Abstract
Toll-like receptors (TLRs) recognize pathogen- and damage-associated molecular patterns and, in turn, trigger the release of cytokines and other immunostimulatory molecules. As a result, TLR agonists are increasingly being investigated as vaccine adjuvants. Many of these agonists are small molecules that quickly diffuse away from the vaccination site, limiting their co-localization with antigens and, thus, their effect. Here, the small-molecule TLR7 agonist 1V209 is conjugated to a positively-charged multidomain peptide (MDP) hydrogel, K2, which was previously shown to act as an adjuvant promoting humoral immunity. Mixing the 1V209-conjugated K2 50:50 with the unfunctionalized K2 produces hydrogels that retain the shear-thinning and self-healing physical properties of the original MDP while improving the solubility of 1V209 more than 200-fold compared to the unconjugated molecule. When co-delivered with ovalbumin as a model antigen, 1V209-functionalized K2 produces a robust Th2 immune response and an antigen-specific Th1 immune response superior to alum, a widely used vaccine adjuvant. Together, these results suggest that K2 MDP hydrogels functionalized with 1V209 are a promising adjuvant for vaccines against infectious diseases, especially those benefiting from a combined Th1 and Th2 immune response.
Collapse
Affiliation(s)
- Erin M Euliano
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Brett H Pogostin
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Anushka Agrawal
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Marina H Yu
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | | | - Tyler P Graf
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Alyssa A Kunkel
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Kiana A Cahue
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Jeffrey D Hartgerink
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX, 77005, USA
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX, 77005, USA
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX, 77005, USA
| |
Collapse
|
2
|
Robang A, Wong KM, Leisen J, Liu R, Radford WL, Rao Sudarshan T, Hudalla GA, Paravastu AK. Parallel β-Sheet Structure and Structural Heterogeneity Detected within Q11 Self-Assembling Peptide Nanofibers. J Phys Chem B 2024; 128:5387-5396. [PMID: 38787393 PMCID: PMC11163420 DOI: 10.1021/acs.jpcb.4c00825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Q11 peptide nanofibers are used as a biomaterial for applications such as antigen presentation and tissue engineering, yet detailed knowledge of molecular-level structure has not been reported. The Q11 peptide sequence was designed using heuristics-based patterning of hydrophobic and polar amino acids with oppositely charged amino acids placed at opposite ends of the sequence to promote antiparallel β-sheet formation. In this work, we employed solid-state nuclear magnetic resonance spectroscopy (NMR) to evaluate whether the molecular organization within Q11 self-assembled peptide nanofibers is consistent with the expectations of the peptide designers. We discovered that Q11 forms a distribution of molecular structures. NMR data from two-dimensional (2D) 13C-13C dipolar-assisted rotational resonance indicate that the K3 and E9 residues between Q11 β-strands are spatially proximate (within ∼0.6 nm). Frequency-selective rotational echo double resonance (fsREDOR) on K3 Nζ and E9 Cδ-labeled sites showed that approximately 9% of the sites are close enough for salt bridge formation to occur. Surprisingly, dipolar recoupling measurements revealed that Q11 peptides do not assemble into antiparallel β-sheets as expected, and structural analysis using Fourier-transform infrared spectroscopy and 2D NMR alone can be misleading. 13C PITHIRDS-CT dipolar recoupling measurements showed that the most abundant structure consists of parallel β-sheets, in contrast to the expected antiparallel β-sheet structure. Structural heterogeneity was detected from 15N{13C} REDOR measurements, with approximately 22% of β-strands having antiparallel nearest neighbors. We cannot propose a complete structural model of Q11 nanofibers because of the complexity involved when examining structurally heterogeneous samples using NMR. Altogether, our results show that while heuristics-based patterning is effective in promoting β-sheet formation, designing a peptide sequence to form a targeted β-strand arrangement remains challenging.
Collapse
Affiliation(s)
- Alicia
S. Robang
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Kong M. Wong
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Johannes Leisen
- School
of Chemistry & Biochemistry, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
| | - Renjie Liu
- J. Crayton
Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Walker L. Radford
- School
of Chemistry & Biochemistry, Georgia
Institute of Technology, Atlanta, Georgia 30332, United States
| | - Tarunya Rao Sudarshan
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Gregory A. Hudalla
- J. Crayton
Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Anant K. Paravastu
- School
of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- Parker
H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
3
|
Rapp PB, Baccile JA, Galimidi RP, Vielmetter J. Engineering Antigen-Specific Tolerance to an Artificial Protein Hydrogel. ACS Biomater Sci Eng 2024; 10:2188-2199. [PMID: 38479351 DOI: 10.1021/acsbiomaterials.3c01430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Artificial protein hydrogels are an emerging class of biomaterials with numerous prospective applications in tissue engineering and regenerative medicine. These materials are likely to be immunogenic due to their frequent incorporation of novel amino acid sequence domains, which often serve a functional role within the material itself. We engineered injectable "self" and "nonself" artificial protein hydrogels, which were predicted to have divergent immune outcomes in vivo on the basis of their primary amino acid sequence. Following implantation in mouse, the nonself gels raised significantly higher antigel antibody titers than the corresponding self gels. Prophylactic administration of a fusion antibody targeting the nonself hydrogel epitopes to DEC-205, an endocytic receptor involved in Treg induction, fully suppressed the elevated antibody titer against the nonself gels. These results suggest that the clinical immune response to artificial protein biomaterials, including those that contain highly antigenic sequence domains, can be tuned through the induction of antigen-specific tolerance.
Collapse
Affiliation(s)
- Peter B Rapp
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, California 91125, United States
| | - Joshua A Baccile
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, California 91125, United States
| | - Rachel P Galimidi
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, California 91125, United States
| | - Jost Vielmetter
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, California 91125, United States
| |
Collapse
|
4
|
Li L, Zheng R, Sun R. Understanding multicomponent low molecular weight gels from gelators to networks. J Adv Res 2024:S2090-1232(24)00126-7. [PMID: 38570015 DOI: 10.1016/j.jare.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/11/2024] [Accepted: 03/29/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND The construction of gels from low molecular weight gelators (LMWG) has been extensively studied in the fields of bio-nanotechnology and other fields. However, the understanding gaps still prevent the prediction of LMWG from the full design of those gel systems. Gels with multicomponent become even more complicated because of the multiple interference effects coexist in the composite gel systems. AIM OF REVIEW This review emphasizes systems view on the understanding of multicomponent low molecular weight gels (MLMWGs), and summarizes recent progress on the construction of desired networks of MLMWGs, including self-sorting and co-assembly, as well as the challenges and approaches to understanding MLMWGs, with the hope that the opportunities from natural products and peptides can speed up the understanding process and close the gaps between the design and prediction of structures. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, understanding the complicated multicomponent gels systems requires a systems perspective on MLMWGs. Secondly, several protocols can be applied to control self-sorting and co-assembly behaviors in those multicomponent gels system, including the certain complementary structures, chirality inducing and dynamic control. Thirdly, the discussion is anchored in challenges and strategies of understanding MLMWGs, and some examples are provided for the understanding of multicomponent gels constructed from small natural products and subtle designed short peptides.
Collapse
Affiliation(s)
- Liangchun Li
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China.
| | - Renlin Zheng
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China
| | - Rongqin Sun
- School of Materials Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China
| |
Collapse
|
5
|
Euliano EM, Pogostin BH, Agrawal A, Yu MH, Baryakova TH, Graf TP, Hartgerink JD, McHugh KJ. A TLR7 Agonist Conjugated to a Nanofibrous Peptide Hydrogel as a Potent Vaccine Adjuvant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583938. [PMID: 38496534 PMCID: PMC10942436 DOI: 10.1101/2024.03.07.583938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Toll-like receptors (TLRs) recognize pathogen- and damage-associated molecular patterns and, in turn, trigger the release of cytokines and other immunostimulatory molecules. As a result, TLR agonists are increasingly being investigated as vaccine adjuvants, though many of these agonists are small molecules that quickly diffuse away from the vaccination site, limiting their co-localization with antigens and, thus, their effect. Here, the small-molecule TLR7 agonist 1V209 is conjugated to a positively-charged multidomain peptide (MDP) hydrogel, K 2 , which was previously shown to act as an adjuvant promoting humoral immunity. Mixing the 1V209-conjugated K 2 50:50 with the unfunctionalized K 2 produces hydrogels that retain the shear-thinning and self-healing physical properties of the original MDP, while improving the solubility of 1V209 more than 200-fold compared to the unconjugated molecule. When co-delivered with ovalbumin as a model antigen, 1V209-functionalized K 2 produces antigen-specific IgG titers that were statistically similar to alum, the gold standard adjuvant, and a significantly lower ratio of Th2-associated IgG1 to Th1-associated IgG2a than alum, suggesting a more balanced Th1 and Th2 response. Together, these results suggest that K 2 MDP hydrogels functionalized with 1V209 are a promising adjuvant for vaccines against infectious diseases, especially those benefiting from a combined Th1 and Th2 immune response. Table of Contents Activation of toll-like receptors (TLRs) stimulates a signaling cascade to induce an immune response. A TLR7 agonist was conjugated to an injectable peptide hydrogel, which was then used to deliver a model vaccine antigen. This platform produced antibody titers similar to the gold standard adjuvant alum and demonstrated an improved balance between Th1- and Th2-mediated immunity over alum.
Collapse
|
6
|
Dodd-O J, Roy A, Siddiqui Z, Jafari R, Coppola F, Ramasamy S, Kolloli A, Kumar D, Kaundal S, Zhao B, Kumar R, Robang AS, Li J, Azizogli AR, Pai V, Acevedo-Jake A, Heffernan C, Lucas A, McShan AC, Paravastu AK, Prasad BVV, Subbian S, Král P, Kumar V. Antiviral fibrils of self-assembled peptides with tunable compositions. Nat Commun 2024; 15:1142. [PMID: 38326301 PMCID: PMC10850501 DOI: 10.1038/s41467-024-45193-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024] Open
Abstract
The lasting threat of viral pandemics necessitates the development of tailorable first-response antivirals with specific but adaptive architectures for treatment of novel viral infections. Here, such an antiviral platform has been developed based on a mixture of hetero-peptides self-assembled into functionalized β-sheets capable of specific multivalent binding to viral protein complexes. One domain of each hetero-peptide is designed to specifically bind to certain viral proteins, while another domain self-assembles into fibrils with epitope binding characteristics determined by the types of peptides and their molar fractions. The self-assembled fibrils maintain enhanced binding to viral protein complexes and retain high resilience to viral mutations. This method is experimentally and computationally tested using short peptides that specifically bind to Spike proteins of SARS-CoV-2. This platform is efficacious, inexpensive, and stable with excellent tolerability.
Collapse
Affiliation(s)
- Joseph Dodd-O
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Roya Jafari
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Francesco Coppola
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Santhamani Ramasamy
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Afsal Kolloli
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Dilip Kumar
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Soni Kaundal
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Boyang Zhao
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ranjeet Kumar
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Alicia S Robang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jeffrey Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Abdul-Rahman Azizogli
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Varun Pai
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Amanda Acevedo-Jake
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Corey Heffernan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
- SAPHTx Inc, Newark, NJ, 07104, USA
| | - Alexandra Lucas
- Center for Personalized Diagnostics and Center for Immunotherapy Vaccines and Virotherapy, Biodesign Institute, Arizona State University, 727 E, Tempe, AZ, USA
| | - Andrew C McShan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Anant K Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - B V Venkataram Prasad
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Petr Král
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Physics, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| | - Vivek Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- SAPHTx Inc, Newark, NJ, 07104, USA.
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, 07103, USA.
| |
Collapse
|
7
|
Ou BS, Baillet J, Picece VCT, Gale EC, Powell AE, Saouaf OM, Yan J, Nejatfard A, Lopez Hernandez H, Appel EA. Nanoparticle-Conjugated Toll-Like Receptor 9 Agonists Improve the Potency, Durability, and Breadth of COVID-19 Vaccines. ACS NANO 2024; 18:3214-3233. [PMID: 38215338 PMCID: PMC10832347 DOI: 10.1021/acsnano.3c09700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Development of effective vaccines for infectious diseases has been one of the most successful global health interventions in history. Though, while ideal subunit vaccines strongly rely on antigen and adjuvant(s) selection, the mode and time scale of exposure to the immune system has often been overlooked. Unfortunately, poor control over the delivery of many adjuvants, which play a key role in enhancing the quality and potency of immune responses, can limit their efficacy and cause off-target toxicities. There is a critical need for improved adjuvant delivery technologies to enhance their efficacy and boost vaccine performance. Nanoparticles have been shown to be ideal carriers for improving antigen delivery due to their shape and size, which mimic viral structures but have been generally less explored for adjuvant delivery. Here, we describe the design of self-assembled poly(ethylene glycol)-b-poly(lactic acid) nanoparticles decorated with CpG, a potent TLR9 agonist, to increase adjuvanticity in COVID-19 vaccines. By controlling the surface density of CpG, we show that intermediate valency is a key factor for TLR9 activation of immune cells. When delivered with the SARS-CoV-2 spike protein, CpG nanoparticle (CpG-NP) adjuvant greatly improves the magnitude and duration of antibody responses when compared to soluble CpG, and results in overall greater breadth of immunity against variants of concern. Moreover, encapsulation of CpG-NP into injectable polymeric-nanoparticle (PNP) hydrogels enhances the spatiotemporal control over codelivery of CpG-NP adjuvant and spike protein antigen such that a single immunization of hydrogel-based vaccines generates humoral responses comparable to those of a typical prime-boost regimen of soluble vaccines. These delivery technologies can potentially reduce the costs and burden of clinical vaccination, both of which are key elements in fighting a pandemic.
Collapse
Affiliation(s)
- Ben S. Ou
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Julie Baillet
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Vittoria C. T.
M. Picece
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
- Department
of Chemistry and Applied Biosciences, ETH
Zurich, Zurich 8093, Switzerland
| | - Emily C. Gale
- Department
of Biochemistry, Stanford University School
of Medicine, Stanford, California 94305, United States
| | - Abigail E. Powell
- Department
of Biochemistry, Stanford University School
of Medicine, Stanford, California 94305, United States
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
| | - Olivia M. Saouaf
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Jerry Yan
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Anahita Nejatfard
- Department
of Biochemistry, Stanford University School
of Medicine, Stanford, California 94305, United States
| | - Hector Lopez Hernandez
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Eric A. Appel
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
- Stanford
ChEM-H, Stanford University, Stanford, California 94305, United States
- Department
of Pediatrics - Endocrinology, Stanford
University School of Medicine, Stanford, California 94305, United States
- Woods
Institute for the Environment, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
8
|
Wang H, Mills J, Sun B, Cui H. Therapeutic Supramolecular Polymers: Designs and Applications. Prog Polym Sci 2024; 148:101769. [PMID: 38188703 PMCID: PMC10769153 DOI: 10.1016/j.progpolymsci.2023.101769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The self-assembly of low-molecular-weight building motifs into supramolecular polymers has unlocked a new realm of materials with distinct properties and tremendous potential for advancing medical practices. Leveraging the reversible and dynamic nature of non-covalent interactions, these supramolecular polymers exhibit inherent responsiveness to their microenvironment, physiological cues, and biomolecular signals, making them uniquely suited for diverse biomedical applications. In this review, we intend to explore the principles of design, synthesis methodologies, and strategic developments that underlie the creation of supramolecular polymers as carriers for therapeutics, contributing to the treatment and prevention of a spectrum of human diseases. We delve into the principles underlying monomer design, emphasizing the pivotal role of non-covalent interactions, directionality, and reversibility. Moreover, we explore the intricate balance between thermodynamics and kinetics in supramolecular polymerization, illuminating strategies for achieving controlled sizes and distributions. Categorically, we examine their exciting biomedical applications: individual polymers as discrete carriers for therapeutics, delving into their interactions with cells, and in vivo dynamics; and supramolecular polymeric hydrogels as injectable depots, with a focus on their roles in cancer immunotherapy, sustained drug release, and regenerative medicine. As the field continues to burgeon, harnessing the unique attributes of therapeutic supramolecular polymers holds the promise of transformative impacts across the biomedical landscape.
Collapse
Affiliation(s)
- Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jason Mills
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Boran Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine, The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
9
|
Tiwari P, Yadav K, Shukla RP, Gautam S, Marwaha D, Sharma M, Mishra PR. Surface modification strategies in translocating nano-vesicles across different barriers and the role of bio-vesicles in improving anticancer therapy. J Control Release 2023; 363:290-348. [PMID: 37714434 DOI: 10.1016/j.jconrel.2023.09.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Nanovesicles and bio-vesicles (BVs) have emerged as promising tools to achieve targeted cancer therapy due to their ability to overcome many of the key challenges currently being faced with conventional chemotherapy. These challenges include the diverse and often complex pathophysiology involving the progression of cancer, as well as the various biological barriers that circumvent therapeutic molecules reaching their target site in optimum concentration. The scientific evidence suggests that surface-functionalized nanovesicles and BVs camouflaged nano-carriers (NCs) both can bypass the established biological barriers and facilitate fourth-generation targeting for the improved regimen of treatment. In this review, we intend to emphasize the role of surface-functionalized nanovesicles and BVs camouflaged NCs through various approaches that lead to an improved internalization to achieve improved and targeted oncotherapy. We have explored various strategies that have been employed to surface-functionalize and biologically modify these vesicles, including the use of biomolecule functionalized target ligands such as peptides, antibodies, and aptamers, as well as the targeting of specific receptors on cancer cells. Further, the utility of BVs, which are made from the membranes of cells such as mesenchymal stem cells (MSCs), white blood cells (WBCs), red blood cells (RBCs), platelets (PLTs) as well as cancer cells also been investigated. Lastly, we have discussed the translational challenges and limitations that these NCs can encounter and still need to be overcome in order to fully realize the potential of nanovesicles and BVs for targeted cancer therapy. The fundamental challenges that currently prevent successful cancer therapy and the necessity of novel delivery systems are in the offing.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
10
|
Shrimali PC, Chen S, Das A, Dreher R, Howard MK, Ryan JJ, Buck J, Kim D, Sprunger ML, Rudra JS, Jackrel ME. Amyloidogenic propensity of self-assembling peptides and their adjuvant potential for use as DNA vaccines. Acta Biomater 2023; 169:464-476. [PMID: 37586449 DOI: 10.1016/j.actbio.2023.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/27/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
De novo designed peptides that self-assemble into cross-β rich fibrillar biomaterials have been pursued as an innovative platform for the development of adjuvant- and inflammation-free vaccines. However, they share structural and morphological properties similar to amyloid species implicated in neurodegenerative diseases, which has been a long-standing concern for their successful translation. Here, we comprehensively characterize the amyloidogenic character of the amphipathic self-assembling cross-β peptide KFE8, compared to pathological amyloid and amyloid-like proteins α-synuclein (α-syn) and TDP-43. Further, we developed plasmid-based DNA vaccines with the KFE8 backbone serving as a scaffold for delivery of a GFP model antigen. We find that expression of tandem repeats of KFE8 is non-toxic and efficiently cleared by autophagy. We also demonstrate that preformed KFE8 fibrils do not cross-seed amyloid formation of α-syn in mammalian cells compared to α-syn preformed fibrils. In mice, vaccination with plasmids encoding the KFE32-GFP fusion protein elicited robust immune responses, inducing production of significantly higher levels of anti-GFP antibodies compared to soluble GFP. Antigen-specific CD8+T cells were also detected in the spleens of vaccinated mice and cytokine profiles from antigen recall assays indicate a balanced Th1/Th2 response. These findings illustrate that cross-β-rich peptide nanofibers have distinct physicochemical properties from those of pathological amyloidogenic proteins, and are an attractive platform for the development of DNA vaccines with self-adjuvanting properties and improved safety profiles. STATEMENT OF SIGNIFICANCE: Biomaterials comprised of self-assembling peptides hold great promise for the development of new vaccines that do not require use of adjuvants. However, these materials have safety concerns, as they self-assemble into cross-β rich fibrils that are structurally similar to amyloid species implicated in disease. Here, we comprehensively study the properties of these biomaterials. We demonstrate that they have distinct properties from pathological proteins. They are non-toxic and do not trigger amyloidogenesis. Vaccination of these materials in mice elicited a robust immune response. Most excitingly, our work suggests that this platform could be used to develop DNA-based vaccines, which have few storage requirements. Further, due to their genetic encoding, longer sequences can be generated and the vaccines will be amenable to modification.
Collapse
Affiliation(s)
- Paresh C Shrimali
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Sheng Chen
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Anirban Das
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA; Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Rachel Dreher
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Matthew K Howard
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Jeremy J Ryan
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Jeremy Buck
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Darren Kim
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Macy L Sprunger
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA
| | - Jai S Rudra
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA.
| | - Meredith E Jackrel
- Department of Chemistry, Washington University, St. Louis, MO 63130, USA.
| |
Collapse
|
11
|
Wu C, Wang M, Sun J, Jia Y, Zhu X, Liu G, Zhu Y, Guan Y, Zhang Z, Pang X. Peptide-drug co-assembling: A potent armament against cancer. Theranostics 2023; 13:5322-5347. [PMID: 37908727 PMCID: PMC10614680 DOI: 10.7150/thno.87356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/19/2023] [Indexed: 11/02/2023] Open
Abstract
Cancer is still one of the major problems threatening human health and the therapeutical efficacies of available treatment choices are often rather low. Due to their favorable biocompatibility, simplicity of modification, and improved therapeutic efficacy, peptide-based self-assembled delivery systems have undergone significant evolution. Physical encapsulation and covalent conjugation are two common approaches to load drugs for peptide assembly-based delivery, which are always associated with drug leaks in the blood circulation system or changed pharmacological activities, respectively. To overcome these difficulties, a more elegant peptide-based assembly strategy is desired. Notably, peptide-mediated co-assembly with drug molecules provides a new method for constructing nanomaterials with improved versatility and structural stability. The co-assembly strategy can be used to design various nanostructures for cancer therapy, such as nanotubes, nanofibrils, hydrogels, and nanovesicles. Recently, these co-assembled nanostructures have gained tremendous attention for their unique superiorities in tumor therapy. This article describes the classification of assembled peptides, driving forces for co-assembly, and specifically, the design methodologies for various drug molecules in co-assembly. It also highlights recent research on peptide-mediated co-assembled delivery systems for cancer therapy. Finally, it summarizes the pros and cons of co-assembly in cancer therapy and offers some suggestions for conquering the challenges in this field.
Collapse
Affiliation(s)
- Can Wu
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Manman Wang
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Jinpan Sun
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Yongyan Jia
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Xiali Zhu
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Gaizhi Liu
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Yanhui Zhu
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Yanbin Guan
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Zhenqiang Zhang
- Academy of Chinese Medicine Science, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| | - Xin Pang
- School of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
12
|
Liang H, Lu Q, Yang J, Yu G. Supramolecular Biomaterials for Cancer Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2023; 6:0211. [PMID: 37705962 PMCID: PMC10496790 DOI: 10.34133/research.0211] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/01/2023] [Indexed: 09/15/2023]
Abstract
Cancer immunotherapy has achieved tremendous successful clinical results and obtained historic victories in tumor treatments. However, great limitations associated with feeble immune responses and serious adverse effects still cannot be neglected due to the complicated multifactorial etiology and pathologic microenvironment in tumors. The rapid development of nanomedical science and material science has facilitated the advanced progress of engineering biomaterials to tackle critical issues. The supramolecular biomaterials with flexible and modular structures have exhibited unparalleled advantages of high cargo-loading efficiency, excellent biocompatibility, and diversiform immunomodulatory activity, thereby providing a powerful weapon for cancer immunotherapy. In past decades, supramolecular biomaterials were extensively explored as versatile delivery platforms for immunotherapeutic agents or designed to interact with the key moleculars in immune system in a precise and controllable manner. In this review, we focused on the crucial role of supramolecular biomaterials in the modulation of pivotal steps during tumor immunotherapy, including antigen delivery and presentation, T lymphocyte activation, tumor-associated macrophage elimination and repolarization, and myeloid-derived suppressor cell depletion. Based on extensive research, we explored the current limitations and development prospects of supramolecular biomaterials in cancer immunotherapy.
Collapse
Affiliation(s)
- Huan Liang
- College of Science,
Nanjing Forestry University, Nanjing 210037, P. R. China
| | - Qingqing Lu
- College of Science,
Nanjing Forestry University, Nanjing 210037, P. R. China
| | - Jie Yang
- College of Science,
Nanjing Forestry University, Nanjing 210037, P. R. China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry,
Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
13
|
Pogostin BH, Yu MH, Azares AR, Euliano EM, Lai CSE, Saenz G, Wu SX, Farsheed AC, Melhorn SM, Graf TP, Woodside DG, Hartgerink JD, McHugh KJ. Multidomain peptide hydrogel adjuvants elicit strong bias towards humoral immunity. Biomater Sci 2022; 10:6217-6229. [PMID: 36102692 PMCID: PMC9717470 DOI: 10.1039/d2bm01242a] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Adjuvants play a critical role in enhancing vaccine efficacy; however, there is a need to develop new immunomodulatory compounds to address emerging pathogens and to expand the use of immunotherapies. Multidomain peptides (MDPs) are materials composed of canonical amino acids that form injectable supramolecular hydrogels under physiological salt and pH conditions. MDP hydrogels are rapidly infiltrated by immune cells in vivo and have previously been shown to influence cytokine production. Therefore, we hypothesized that these immunostimulatory characteristics would allow MDPs to function as vaccine adjuvants. Herein, we demonstrate that loading antigen into MDP hydrogels does not interfere with their rheological properties and that positively charged MDPs can act as antigen depots, as demonstrated by their ability to release ovalbumin (OVA) over a period of 7-9 days in vivo. Mice vaccinated with MDP-adjuvanted antigen generated significantly higher IgG titers than mice treated with the unadjuvanted control, suggesting that these hydrogels potentiate humoral immunity. Interestingly, MDP hydrogels did not elicit a robust cellular immune response, as indicated by the lower production of IgG2c and smaller populations of tetramer-positive CD8+ T splenocytes compared to mice vaccinated alum-adjuvanted OVA. Together, the data suggest that MDP hydrogel adjuvants strongly bias the immune response towards humoral immunity while evoking a very limited cellular immune response. As a result, MDPs may have the potential to serve as adjuvants for applications that benefit exclusively from humoral immunity.
Collapse
Affiliation(s)
- Brett H Pogostin
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Marina H Yu
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Alon R Azares
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, TX, 77030, USA
| | - Erin M Euliano
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | | | - Gabriel Saenz
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Samuel X Wu
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Adam C Farsheed
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Sarah M Melhorn
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Tyler P Graf
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| | - Darren G Woodside
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, TX, 77030, USA
| | - Jeffrey D Hartgerink
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, Houston, TX, 77005, USA.
| |
Collapse
|
14
|
Chen JL, Fries CN, Berendam SJ, Rodgers NS, Roe EF, Wu Y, Li SH, Jain R, Watts B, Eudailey J, Barfield R, Chan C, Moody MA, Saunders KO, Pollara J, Permar SR, Collier JH, Fouda GG. Self-assembling peptide nanofiber HIV vaccine elicits robust vaccine-induced antibody functions and modulates Fc glycosylation. SCIENCE ADVANCES 2022; 8:eabq0273. [PMID: 36149967 PMCID: PMC9506727 DOI: 10.1126/sciadv.abq0273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 08/04/2022] [Indexed: 06/16/2023]
Abstract
To develop vaccines for certain key global pathogens such as HIV, it is crucial to elicit both neutralizing and non-neutralizing Fc-mediated effector antibody functions. Clinical evidence indicates that non-neutralizing antibody functions including antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) contribute to protection against several pathogens. In this study, we demonstrated that conjugation of HIV Envelope (Env) antigen gp120 to a self-assembling nanofiber material named Q11 induced antibodies with higher breadth and functionality when compared to soluble gp120. Immunization with Q11-conjugated gp120 vaccine (gp120-Q11) demonstrated higher tier 1 neutralization, ADCP, and ADCC as compared to soluble gp120. Moreover, Q11 conjugation altered the Fc N-glycosylation profile of antigen-specific antibodies, leading to a phenotype associated with increased ADCC in animals immunized with gp120-Q11. Thus, this nanomaterial vaccine strategy can enhance non-neutralizing antibody functions possibly through modulation of immunoglobulin G Fc N-glycosylation.
Collapse
Affiliation(s)
- Jui-Lin Chen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chelsea N. Fries
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Stella J. Berendam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nicole S. Rodgers
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Emily F. Roe
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Shuk Hang Li
- The Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rishabh Jain
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Brian Watts
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Joshua Eudailey
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham NC 27710, USA
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, NC 27707, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham NC 27710, USA
- Center for Human Systems Immunology, Duke University School of Medicine, Durham, NC 27707, USA
| | - M. Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin O. Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Justin Pollara
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joel H. Collier
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
15
|
Mañas‐Torres MC, Illescas‐Lopez S, Gavira JA, de Cienfuegos LÁ, Marchesan S. Interactions Between Peptide Assemblies and Proteins for Medicine. Isr J Chem 2022. [DOI: 10.1002/ijch.202200018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Mari C. Mañas‐Torres
- Departamento de Química Orgánica, Facultad de Ciencias Unidad de Excelencia Química Aplicada a Biomedicina y Medioambiente (UEQ) Universidad de Granada, (UGR) C. U. Fuentenueva Avda. Severo Ochoa s/n E-18071 Granada
| | - Sara Illescas‐Lopez
- Departamento de Química Orgánica, Facultad de Ciencias Unidad de Excelencia Química Aplicada a Biomedicina y Medioambiente (UEQ) Universidad de Granada, (UGR) C. U. Fuentenueva Avda. Severo Ochoa s/n E-18071 Granada
| | - José A. Gavira
- Laboratorio de Estudios Cristalográficos Instituto Andaluz de Ciencias de la Tierra (Consejo Superior de Investigaciones Científicas-UGR) Avenida de las Palmeras 4 18100 Armilla, UEQ Granada Spain
| | - Luis Álvarez de Cienfuegos
- Departamento de Química Orgánica, Facultad de Ciencias Unidad de Excelencia Química Aplicada a Biomedicina y Medioambiente (UEQ) Universidad de Granada, (UGR) C. U. Fuentenueva Avda. Severo Ochoa s/n E-18071 Granada
- Instituto de Investigación Biosanitaria ibs Granada Spain
| | - Silvia Marchesan
- Chemical and Pharmaceutical Sciences Department University of Trieste Via L. Giorgieri 1 Trieste 34127 Italy
| |
Collapse
|
16
|
Files MA, Naqvi KF, Saito TB, Clover TM, Rudra JS, Endsley JJ. Self-adjuvanting nanovaccines boost lung-resident CD4 + T cell immune responses in BCG-primed mice. NPJ Vaccines 2022; 7:48. [PMID: 35474079 PMCID: PMC9043212 DOI: 10.1038/s41541-022-00466-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/21/2022] [Indexed: 01/04/2023] Open
Abstract
Heterologous vaccine regimens could extend waning protection in the global population immunized with Mycobacterium bovis Bacille Calmette-Guerin (BCG). We demonstrate that pulmonary delivery of peptide nanofibers (PNFs) bearing an Ag85B CD4+ T cell epitope increased the frequency of antigen-specific T cells in BCG-primed mice, including heterogenous populations with tissue resident memory (Trm) and effector memory (Tem) phenotype, and functional cytokine recall. Adoptive transfer of dendritic cells pulsed with Ag85B-bearing PNFs further expanded the frequency and functional repertoire of memory CD4+ T cells. Transcriptomic analysis suggested that the adjuvanticity of peptide nanofibers is, in part, due to the release of damage-associated molecular patterns. A single boost with monovalent Ag85B PNF in BCG-primed mice did not reduce lung bacterial burden compared to BCG alone following aerosol Mtb challenge. These findings support the need for novel BCG booster strategies that activate pools of Trm cells with potentially diverse localization, trafficking, and immune function.
Collapse
Grants
- R01 AI130278 NIAID NIH HHS
- R21 AI115302 NIAID NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases (NIAID)
- Predoctoral Fellowship, Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas 77555
- Predoctoral Fellowship, James W. McLaughlin Endowment, University of Texas Medical Branch, Galveston, Texas, 77555
- Washington University McKelvey School of Engineering, Department of Biomedical Engineering Commitment Funds (12-360-94361J)
Collapse
Affiliation(s)
- Megan A Files
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Institute of Translational Science, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kubra F Naqvi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tais B Saito
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Tara M Clover
- Comprehensive Industrial Hygiene Laboratory (CIHL), Navy Environmental and Preventive Medicine Unit TWO (NEPMU-2), Department of the Navy, Norfolk, VA, 23551, USA
| | - Jai S Rudra
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA.
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
17
|
Aburayan WS, Alajmi AM, Alfahad AJ, Alsharif WK, Alshehri AA, Booq RY, Alsudir SA, Alsulaihem FM, Bukhary HA, Badr MY, Alyamani EJ, Tawfik EA. Melittin from Bee Venom Encapsulating Electrospun Fibers as a Potential Antimicrobial Wound Dressing Patches for Skin Infections. Pharmaceutics 2022; 14:pharmaceutics14040725. [PMID: 35456558 PMCID: PMC9030956 DOI: 10.3390/pharmaceutics14040725] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Skin infection compromises the body’s natural defenses. Several antibiotics are no longer effective owing to the evolution of antimicrobial-resistant (AMR) bacteria, hence, the constant development of novel antibacterial agents. Naturally occurring antibacterial agents may be potential candidates for AMR bacterial infection treatments; however, caution should be taken when administering such agents due to the high incidence of toxicity. A fibrous material system from a biocompatible polymer that could be used as a skin patch for skin infections treatment caused by AMR bacteria is proposed in this study. Bee venom’s active ingredient, melittin, was fabricated using electrospinning technology. Scanning electron microscopy showed that melittin-loaded fibers had smooth surfaces with no signs of beads or pores. The average diameter of this fibrous system was measured to be 1030 ± 160 nm, indicating its successful preparation. The melittin fibers’ drug loading and entrapment efficiency (EE%) were 49 ± 3 µg/mg and 84 ± 5%, respectively. This high EE% can be another successful preparatory criterion. An in vitro release study demonstrated that 40% of melittin was released after 5 min and achieved complete release after 120 min owing to the hydrophilic nature of the PVP polymer. A concentration of ≤10 µg/mL was shown to be safe for use on human dermal fibroblasts HFF-1 after 24-h exposure, while an antibacterial MIC study found that 5 μg/mL was the effective antimicrobial concentration for S. aureus, A. baumannii, E. coli and Candida albicans yeast. A melittin-loaded fibrous system demonstrated an antibacterial zone of inhibition equivalent to the control (melittin discs), suggesting its potential use as a wound dressing patch for skin infections.
Collapse
Affiliation(s)
- Walaa S. Aburayan
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Areej M. Alajmi
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Ahmed J. Alfahad
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Wijdan K. Alsharif
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Abdullah A. Alshehri
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Rayan Y. Booq
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Samar A. Alsudir
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Fatemah M. Alsulaihem
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Haitham A. Bukhary
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 24381, Saudi Arabia; (H.A.B.); (M.Y.B.)
| | - Moutaz Y. Badr
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 24381, Saudi Arabia; (H.A.B.); (M.Y.B.)
| | - Essam J. Alyamani
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
| | - Essam A. Tawfik
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (W.S.A.); (A.M.A.); (A.J.A.); (W.K.A.); (A.A.A.); (R.Y.B.); (S.A.A.); (F.M.A.); (E.J.A.)
- Correspondence:
| |
Collapse
|
18
|
Gray VP, Amelung CD, Duti IJ, Laudermilch EG, Letteri RA, Lampe KJ. Biomaterials via peptide assembly: Design, characterization, and application in tissue engineering. Acta Biomater 2022; 140:43-75. [PMID: 34710626 PMCID: PMC8829437 DOI: 10.1016/j.actbio.2021.10.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/23/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
A core challenge in biomaterials, with both fundamental significance and technological relevance, concerns the rational design of bioactive microenvironments. Designed properly, peptides can undergo supramolecular assembly into dynamic, physical hydrogels that mimic the mechanical, topological, and biochemical features of native tissue microenvironments. The relatively facile, inexpensive, and automatable preparation of peptides, coupled with low batch-to-batch variability, motivates the expanded use of assembling peptide hydrogels for biomedical applications. Integral to realizing dynamic peptide assemblies as functional biomaterials for tissue engineering is an understanding of the molecular and macroscopic features that govern assembly, morphology, and biological interactions. In this review, we first discuss the design of assembling peptides, including primary structure (sequence), secondary structure (e.g., α-helix and β-sheets), and molecular interactions that facilitate assembly into multiscale materials with desired properties. Next, we describe characterization tools for elucidating molecular structure and interactions, morphology, bulk properties, and biological functionality. Understanding of these characterization methods enables researchers to access a variety of approaches in this ever-expanding field. Finally, we discuss the biological properties and applications of peptide-based biomaterials for engineering several important tissues. By connecting molecular features and mechanisms of assembling peptides to the material and biological properties, we aim to guide the design and characterization of peptide-based biomaterials for tissue engineering and regenerative medicine. STATEMENT OF SIGNIFICANCE: Engineering peptide-based biomaterials that mimic the topological and mechanical properties of natural extracellular matrices provide excellent opportunities to direct cell behavior for regenerative medicine and tissue engineering. Here we review the molecular-scale features of assembling peptides that result in biomaterials that exhibit a variety of relevant extracellular matrix-mimetic properties and promote beneficial cell-biomaterial interactions. Aiming to inspire and guide researchers approaching this challenge from both the peptide biomaterial design and tissue engineering perspectives, we also present characterization tools for understanding the connection between peptide structure and properties and highlight the use of peptide-based biomaterials in neural, orthopedic, cardiac, muscular, and immune engineering applications.
Collapse
Affiliation(s)
- Vincent P Gray
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Connor D Amelung
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Israt Jahan Duti
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Emma G Laudermilch
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States
| | - Rachel A Letteri
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903, United States.
| |
Collapse
|
19
|
Pogostin BH, McHugh KJ. Novel Vaccine Adjuvants as Key Tools for Improving Pandemic Preparedness. Bioengineering (Basel) 2021; 8:155. [PMID: 34821721 PMCID: PMC8615241 DOI: 10.3390/bioengineering8110155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
Future infectious disease outbreaks are inevitable; therefore, it is critical that we maximize our readiness for these events by preparing effective public health policies and healthcare innovations. Although we do not know the nature of future pathogens, antigen-agnostic platforms have the potential to be broadly useful in the rapid response to an emerging infection-particularly in the case of vaccines. During the current COVID-19 pandemic, recent advances in mRNA engineering have proven paramount in the rapid design and production of effective vaccines. Comparatively, however, the development of new adjuvants capable of enhancing vaccine efficacy has been lagging. Despite massive improvements in our understanding of immunology, fewer than ten adjuvants have been approved for human use in the century since the discovery of the first adjuvant. Modern adjuvants can improve vaccines against future pathogens by reducing cost, improving antigen immunogenicity, and increasing antigen stability. In this perspective, we survey the current state of adjuvant use, highlight potentially impactful preclinical adjuvants, and propose new measures to accelerate adjuvant safety testing and technology sharing to enable the use of "off-the-shelf" adjuvant platforms for rapid vaccine testing and deployment in the face of future pandemics.
Collapse
Affiliation(s)
| | - Kevin J. McHugh
- Department of Bioengineering, Rice University, Houston, TX 77030, USA;
| |
Collapse
|
20
|
O'Neill CL, Shrimali PC, Clapacs ZE, Files MA, Rudra JS. Peptide-based supramolecular vaccine systems. Acta Biomater 2021; 133:153-167. [PMID: 34010691 PMCID: PMC8497425 DOI: 10.1016/j.actbio.2021.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022]
Abstract
Currently approved replication-competent and inactivated vaccines are limited by excessive reactogenicity and poor safety profiles, while subunit vaccines are often insufficiently immunogenic without co-administering exogenous adjuvants. Self-assembling peptide-, peptidomimetic-, and protein-based biomaterials offer a means to overcome these challenges through their inherent modularity, multivalency, and biocompatibility. As these scaffolds are biologically derived and present antigenic arrays reminiscent of natural viruses, they are prone to immune recognition and are uniquely capable of functioning as self-adjuvanting vaccine delivery vehicles that improve humoral and cellular responses. Beyond this intrinsic immunological advantage, the wide range of available amino acids allows for facile de novo design or straightforward modifications to existing sequences. This has permitted the development of vaccines and immunotherapies tailored to specific disease models, as well as generalizable platforms that have been successfully applied to prevent or treat numerous infectious and non-infectious diseases. In this review, we briefly introduce the immune system, discuss the structural determinants of coiled coils, β-sheets, peptide amphiphiles, and protein subunit nanoparticles, and highlight the utility of these materials using notable examples of their innate and adaptive immunomodulatory capacity. STATEMENT OF SIGNIFICANCE: Subunit vaccines have recently gained considerable attention due to their favorable safety profiles relative to traditional whole-cell vaccines; however, their reduced efficacy requires co-administration of reactogenic adjuvants to boost immune responses. This has led to collaborative efforts between engineers and immunologists to develop nanomaterial-based vaccination platforms that can elicit protection without deleterious side effects. Self-assembling peptidic biomaterials are a particularly attractive approach to this problem, as their structure and function can be controlled through primary sequence design and their capacity for multivalent presentation of antigens grants them intrinsic self-adjuvanticity. This review introduces the various architectures adopted by self-assembling peptides and discusses their application as modulators of innate and adaptive immunity.
Collapse
Affiliation(s)
- Conor L O'Neill
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Paresh C Shrimali
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Zoe E Clapacs
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| | - Megan A Files
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States.
| | - Jai S Rudra
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States.
| |
Collapse
|
21
|
Fries CN, Chen JL, Dennis ML, Votaw NL, Eudailey J, Watts BE, Hainline KM, Cain DW, Barfield R, Chan C, Moody MA, Haynes BF, Saunders KO, Permar SR, Fouda GG, Collier JH. HIV envelope antigen valency on peptide nanofibers modulates antibody magnitude and binding breadth. Sci Rep 2021; 11:14494. [PMID: 34262096 PMCID: PMC8280189 DOI: 10.1038/s41598-021-93702-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/23/2021] [Indexed: 01/02/2023] Open
Abstract
A major challenge in developing an effective vaccine against HIV-1 is the genetic diversity of its viral envelope. Because of the broad range of sequences exhibited by HIV-1 strains, protective antibodies must be able to bind and neutralize a widely mutated viral envelope protein. No vaccine has yet been designed which induces broadly neutralizing or protective immune responses against HIV in humans. Nanomaterial-based vaccines have shown the ability to generate antibody and cellular immune responses of increased breadth and neutralization potency. Thus, we have developed supramolecular nanofiber-based immunogens bearing the HIV gp120 envelope glycoprotein. These immunogens generated antibody responses that had increased magnitude and binding breadth compared to soluble gp120. By varying gp120 density on nanofibers, we determined that increased antigen valency was associated with increased antibody magnitude and germinal center responses. This study presents a proof-of-concept for a nanofiber vaccine platform generating broad, high binding antibody responses against the HIV-1 envelope glycoprotein.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Maria L Dennis
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Nicole L Votaw
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Joshua Eudailey
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Brian E Watts
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kelly M Hainline
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA
| | - Derek W Cain
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kevin O Saunders
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Pediatrics, New York-Presbyterian/Weill Cornell Medicine, New York, NY, 10065, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Pediatrics, Duke University Medical Center, Duke University School of Medicine, Box 103020, Durham, NC, 27710, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, 101 Science Dr., Campus, Box 90281, Durham, NC, 27708, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
22
|
Zhang Z, Ai S, Yang Z, Li X. Peptide-based supramolecular hydrogels for local drug delivery. Adv Drug Deliv Rev 2021; 174:482-503. [PMID: 34015417 DOI: 10.1016/j.addr.2021.05.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
Peptide-based supramolecular hydrogels have shown great promise as drug delivery systems (DDSs) because of their excellent biocompatibility, biodegradability, biological function, synthetic feasibility, and responsiveness to external stimuli. Self-assembling peptide molecules are able rationally designed into specific nanoarchitectures in response to the different environmental factors under different circumstances. Among all stimuli that have been investigated, utilizing inherent biological microenvironment, such as metal ions, enzymes and endogenous redox species, to trigger self-assembly endows such systems spatiotemporal controllability to transport therapeutics more accurately. Materials formed by weak non-covalent interactions result in the shear-thinning and immediate recovery behavior. Thus, they are injectable via a syringe or catheter, making them the ideal vehicles to deliver drugs. Based on the above merits, self-assembling peptide-based DDSs have been applied to treat various diseases via direct administration at the lesion site. Herein, in this review, we outline the triggers for inducing peptide-based hydrogels formation and serving as DDSs. We also described the advancements of peptide-based supramolecular hydrogels for local drug delivery, including intratumoral, subcutaneous, ischemia-related tissue (intramyocardial, intrarenal, and ischemic hind limb), and ocular administration. Finally, we give a brief perspective about the prospects and challenges in this field.
Collapse
Affiliation(s)
- Zhenghao Zhang
- Institute of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, PR China
| | - Sifan Ai
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China.
| | - Xingyi Li
- Institute of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, PR China.
| |
Collapse
|
23
|
Zuo R, Liu R, Olguin J, Hudalla GA. Glycosylation of a Nonfibrillizing Appendage Alters the Self-Assembly Pathway of a Synthetic β-Sheet Fibrillizing Peptide. J Phys Chem B 2021; 125:6559-6571. [PMID: 34128680 PMCID: PMC9191660 DOI: 10.1021/acs.jpcb.1c02083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Owing to their biocompatibility and biodegradability, short synthetic peptides that self-assemble into elongated β-sheet fibers (i.e., peptide nanofibers) are widely used to create biomaterials for diverse medical and biotechnology applications. Glycosylation, which is a common protein post-translational modification, is gaining interest for creating peptide nanofibers that can mimic the function of natural carbohydrate-modified proteins. Recent reports have shown that glycosylation can disrupt the fibrillization of natural amyloid-forming peptides. Here, using transmission electron microscopy, fluorescence microscopy, and thioflavin T spectroscopy, we show that glycosylation at a site external to the fibrillization domain can alter the self-assembly pathway of a synthetic fibrillizing peptide, NSGSGQQKFQFQFEQQ (NQ11). Specifically, an NQ11 variant modified with N-linked N-acetylglucosamine, N(GlcNAc)SGSG-Q11 (GQ11), formed β-sheet nanofibers more slowly than NQ11 in deionized water (pH 5.8), which correlated to the tendency of GQ11 to form a combination of short fibrils and nonfibrillar aggregates, whereas NQ11 formed extended nanofibers. Acidic phosphate buffer slowed the rate of GQ11 fibrillization and altered the morphology of the structures formed yet had no effect on NQ11 fibrillization rate or morphology. The buffer ionic strength had no effect on the fibrillization rate of either peptide, while the diphosphate anion had a similar effect on the rate of fibrillization of both peptides. Collectively, these data demonstrate that a glycan moiety located external to the β-sheet fibrillizing domain can alter the pH-dependent self-assembly pathway of a synthetic peptide, leading to significant changes in the fibril mass and morphology of the structures formed. These observations add to the understanding of the effect of glycosylation on peptide self-assembly and should guide future efforts to develop biomaterials from synthetic β-sheet fibrillizing glycopeptides.
Collapse
Affiliation(s)
- Ran Zuo
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Juanpablo Olguin
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Gregory A. Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
24
|
Roth G, Saouaf OM, Smith AAA, Gale EC, Hernández MA, Idoyaga J, Appel EA. Prolonged Codelivery of Hemagglutinin and a TLR7/8 Agonist in a Supramolecular Polymer-Nanoparticle Hydrogel Enhances Potency and Breadth of Influenza Vaccination. ACS Biomater Sci Eng 2021; 7:1889-1899. [PMID: 33404236 PMCID: PMC8153386 DOI: 10.1021/acsbiomaterials.0c01496] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022]
Abstract
The sustained release of vaccine cargo has been shown to improve humoral immune responses to challenging pathogens such as influenza. Extended codelivery of antigen and adjuvant prolongs germinal center reactions, thus improving antibody affinity maturation and the ability to neutralize the target pathogen. Here, we develop an injectable, physically cross-linked polymer-nanoparticle (PNP) hydrogel system to prolong the local codelivery of hemagglutinin and a toll-like receptor 7/8 agonist (TLR7/8a) adjuvant. By tethering the TLR7/8a to a NP motif within the hydrogels (TLR7/8a-NP), the dynamic mesh of the PNP hydrogels enables codiffusion of the adjuvant and protein antigen (hemagglutinin), therefore enabling sustained codelivery of these two physicochemically distinct molecules. We show that subcutaneous delivery of PNP hydrogels carrying hemagglutinin and TLR7/8a-NP in mice improves the magnitude and duration of antibody titers in response to a single injection vaccination compared to clinically used adjuvants. Furthermore, the PNP gel-based slow delivery of influenza vaccines led to increased breadth of antibody responses against future influenza variants, including a future pandemic variant, compared to clinical adjuvants. In summary, this work introduces a simple and effective vaccine delivery platform that increases the potency and durability of influenza subunit vaccines.
Collapse
Affiliation(s)
- Gillie
A. Roth
- Department
of Bioengineering, Stanford University, 443 Via Ortega, Stanford, California 94305, United States
| | - Olivia M. Saouaf
- Department
of Materials Science & Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Anton A. A. Smith
- Department
of Materials Science & Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Emily C. Gale
- Department
of Biochemistry, Stanford University School
of Medicine, 279 Campus Drive, Stanford, California 94305, United States
| | - Marcela Alcántara Hernández
- Department
of Microbiology & Immunology, Stanford
University School of Medicine, 299 Campus Drive, Stanford, California 94305, United States
- Program
in Immunology, Stanford University School
of Medicine, 240 Pasteur Drive, Stanford, California 94305, United States
| | - Juliana Idoyaga
- Department
of Microbiology & Immunology, Stanford
University School of Medicine, 299 Campus Drive, Stanford, California 94305, United States
- Program
in Immunology, Stanford University School
of Medicine, 240 Pasteur Drive, Stanford, California 94305, United States
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, 240 Pasteur Drive, Stanford, California 94305, United States
- ChEM-H
Institute, Stanford University, 290 Jane Stanford Way, Stanford, California 94305, United States
| | - Eric A. Appel
- Department
of Bioengineering, Stanford University, 443 Via Ortega, Stanford, California 94305, United States
- Department
of Materials Science & Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, 240 Pasteur Drive, Stanford, California 94305, United States
- ChEM-H
Institute, Stanford University, 290 Jane Stanford Way, Stanford, California 94305, United States
- Department
of Pediatrics - Endocrinology, Stanford
University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, United States
| |
Collapse
|
25
|
Webber MJ, Pashuck ET. (Macro)molecular self-assembly for hydrogel drug delivery. Adv Drug Deliv Rev 2021; 172:275-295. [PMID: 33450330 PMCID: PMC8107146 DOI: 10.1016/j.addr.2021.01.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 01/15/2023]
Abstract
Hydrogels prepared via self-assembly offer scalable and tunable platforms for drug delivery applications. Molecular-scale self-assembly leverages an interplay of attractive and repulsive forces; drugs and other active molecules can be incorporated into such materials by partitioning in hydrophobic domains, affinity-mediated binding, or covalent integration. Peptides have been widely used as building blocks for self-assembly due to facile synthesis, ease of modification with bioactive molecules, and precise molecular-scale control over material properties through tunable interactions. Additional opportunities are manifest in stimuli-responsive self-assembly for more precise drug action. Hydrogels can likewise be fabricated from macromolecular self-assembly, with both synthetic polymers and biopolymers used to prepare materials with controlled mechanical properties and tunable drug release. These include clinical approaches for solubilization and delivery of hydrophobic drugs. To further enhance mechanical properties of hydrogels prepared through self-assembly, recent work has integrated self-assembly motifs with polymeric networks. For example, double-network hydrogels capture the beneficial properties of both self-assembled and covalent networks. The expanding ability to fabricate complex and precise materials, coupled with an improved understanding of biology, will lead to new classes of hydrogels specifically tailored for drug delivery applications.
Collapse
Affiliation(s)
- Matthew J Webber
- University of Notre Dame, Department of Chemical & Biomolecular Engineering, Notre Dame, IN 46556, USA.
| | - E Thomas Pashuck
- Lehigh University, Department of Bioengineering, Bethlehem, PA 18015, USA.
| |
Collapse
|
26
|
Liu M, Wang Z, Feng D, Shang Y, Li X, Liu J, Li C, Yang Z. An Insulin-Inspired Supramolecular Hydrogel for Prevention of Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003599. [PMID: 34026440 PMCID: PMC8132061 DOI: 10.1002/advs.202003599] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/11/2021] [Indexed: 05/10/2023]
Abstract
Supramolecular peptide hydrogel has shown promising potential in vaccine development largely because of its ability to function both as antigen depot and immune adjuvant. Nap-GdFdFdY, a tetrapeptide hydrogel that has been previously reported to exhibit adjuvant effect, is inadvertently found to contain conserved peptide sequence for insulin, proinsulin, and glutamic acid decarboxylase, 3 major autoantigens for the autoimmune type 1 diabetes (T1D). At present, despite being managed clinically with insulin replacement therapy, T1D remains a major health threat with rapidly increasing incidences, especially in children and young adults, and antigen-specific immune tolerance induction has been proposed as a feasible approach to prevent or delay T1D progression at an early stage. Here, it is reported that innoculation of Nap-GdFdFdY leads to complete protection of nonobese diabetic (NOD) mice from T1D development till the age of 36 weeks. Better maintenance of pancreatic islet morphology with minimal immune cell infiltration is also observed from mice exposed to Nap-GdFdFdY. This beneficial impact is mainly due to its facilitative role on enhancing peripheral T regulatory cell (Treg) population, shown as increased splenic Treg percentage, and function, demonstrated by maintenance of circulating TGF-β1 level. Serum cytokine microarray data further implicate a "buffering" role of Nap-GdFdFdY on systemic inflammatory tone in NOD mice. Thus, with its versatility, applicability, and excellent potency, Nap-GdFdFdY is posited as a novel therapeutic intervention for T1D.
Collapse
Affiliation(s)
- Mohan Liu
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhongyan Wang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Dandan Feng
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Yuna Shang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
| | - Xinxin Li
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Chen Li
- Tianjin Key Laboratory of Biomedical MaterialsBiomedical Barriers Research CentreInstitute of Biomedical EngineeringChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhimou Yang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyCollaborative Innovation Centre of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071P. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu221004P. R. China
| |
Collapse
|
27
|
Modular complement assemblies for mitigating inflammatory conditions. Proc Natl Acad Sci U S A 2021; 118:2018627118. [PMID: 33876753 DOI: 10.1073/pnas.2018627118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Complement protein C3dg, a key linkage between innate and adaptive immunity, is capable of stimulating both humoral and cell-mediated immune responses, leading to considerable interest in its use as a molecular adjuvant. However, the potential of C3dg as an adjuvant is limited without ways of controllably assembling multiple copies of it into vaccine platforms. Here, we report a strategy to assemble C3dg into supramolecular nanofibers with excellent compositional control, using β-tail fusion tags. These assemblies were investigated as therapeutic active immunotherapies, which may offer advantages over existing biologics, particularly toward chronic inflammatory diseases. Supramolecular assemblies based on the Q11 peptide system containing β-tail-tagged C3dg, B cell epitopes from TNF, and the universal T cell epitope PADRE raised strong antibody responses against both TNF and C3dg, and prophylactic immunization with these materials significantly improved protection in a lethal TNF-mediated inflammation model. Additionally, in a murine model of psoriasis induced by imiquimod, the C3dg-adjuvanted nanofiber vaccine performed as well as anti-TNF monoclonal antibodies. Nanofibers containing only β-tail-C3dg and lacking the TNF B cell epitope also showed improvements in both models, suggesting that supramolecular C3dg, by itself, played an important therapeutic role. We observed that immunization with β-tail-C3dg caused the expansion of an autoreactive C3dg-specific T cell population, which may act to dampen the immune response, preventing excessive inflammation. These findings indicate that molecular assemblies displaying C3dg warrant further development as active immunotherapies.
Collapse
|
28
|
Fries CN, Curvino EJ, Chen JL, Permar SR, Fouda GG, Collier JH. Advances in nanomaterial vaccine strategies to address infectious diseases impacting global health. NATURE NANOTECHNOLOGY 2021; 16:1-14. [PMID: 32807876 DOI: 10.1038/s41565-020-0739-9] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 06/23/2020] [Indexed: 05/18/2023]
Abstract
Despite the overwhelming success of vaccines in preventing infectious diseases, there remain numerous globally devastating diseases without fully protective vaccines, particularly human immunodeficiency virus (HIV), malaria and tuberculosis. Nanotechnology approaches are being developed both to design new vaccines against these diseases as well as to facilitate their global implementation. The reasons why a given pathogen may present difficulties for vaccine design are unique and tied to the co-evolutionary history of the pathogen and humans, but there are common challenges that nanotechnology is beginning to help address. In each case, a successful vaccine will need to raise immune responses that differ from the immune responses raised by normal infection. Nanomaterials, with their defined compositions, commonly modular construction, and length scales allowing the engagement of key immune pathways, collectively facilitate the iterative design process necessary to identify such protective immune responses and achieve them reliably. Nanomaterials also provide strategies for engineering the trafficking and delivery of vaccine components to key immune cells and lymphoid tissues, and they can be highly multivalent, improving their engagement with the immune system. This Review will discuss these aspects along with recent nanomaterial advances towards vaccines against infectious disease, with a particular emphasis on HIV/AIDS, malaria and tuberculosis.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
29
|
Thota C, Mikolajczak DJ, Roth C, Koksch B. Enhancing Antimicrobial Peptide Potency through Multivalent Presentation on Coiled-Coil Nanofibrils. ACS Med Chem Lett 2021; 12:67-73. [PMID: 33488966 PMCID: PMC7812673 DOI: 10.1021/acsmedchemlett.0c00425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/08/2020] [Indexed: 12/22/2022] Open
Abstract
Antibiotic-resistant microbes have become a global health threat. New delivery systems that enhance the efficacy of antibiotics and/or overcome the resistances can help combat them. In this context, we present FF03, a fibril-forming α-helical coiled-coil peptide that functions as an efficient scaffold for the multivalent presentation of the weakly cationic antimicrobial peptide (AMP) IN4. The resulting IN4-decorated FF03 coiled-coil fibrils (FF03 + IN4) are nonhemolytic and noncytotoxic and show enhanced antimicrobial activity relative to unconjugated IN4 and standard antibiotics against several bacterial strains. Scanning electron microscopy analysis shows that FF03 + IN4 nanofibers disrupt methicillin-resistant Staphylococcus aureus membranes, indicating a surface-level mode of action. Furthermore, transmission electron microscopy and circular dichroism studies indicate that decoration of the FF03 scaffold with IN4 does not alter the secondary-structure propensity or fibril-forming properties of FF03. Thus, the approach reported herein provides a new peptidic scaffold for the multivalent presentation of AMPs to obtain nonhemolytic and noncytotoxic antimicrobial systems with improved efficacy relative to the unconjugated AMP analogues.
Collapse
Affiliation(s)
- Chaitanya
Kumar Thota
- Department
of Chemistry and Biochemistry, Freie Universität
Berlin, Takustrasse 3, 14195 Berlin, Germany
| | - Dorian J. Mikolajczak
- Department
of Chemistry and Biochemistry, Freie Universität
Berlin, Takustrasse 3, 14195 Berlin, Germany
| | - Christian Roth
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, 14195 Berlin, Germany
| | - Beate Koksch
- Department
of Chemistry and Biochemistry, Freie Universität
Berlin, Takustrasse 3, 14195 Berlin, Germany
| |
Collapse
|
30
|
Seroski DT, Dong X, Wong KM, Liu R, Shao Q, Paravastu AK, Hall CK, Hudalla GA. Charge guides pathway selection in β-sheet fibrillizing peptide co-assembly. Commun Chem 2020; 3:172. [PMID: 36703436 PMCID: PMC9814569 DOI: 10.1038/s42004-020-00414-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/15/2020] [Indexed: 01/29/2023] Open
Abstract
Peptide co-assembly is attractive for creating biomaterials with new forms and functions. Emergence of these properties depends on the peptide content of the final assembled structure, which is difficult to predict in multicomponent systems. Here using experiments and simulations we show that charge governs content by affecting propensity for self- and co-association in binary CATCH(+/-) peptide systems. Equimolar mixtures of CATCH(2+/2-), CATCH(4+/4-), and CATCH(6+/6-) formed two-component β-sheets. Solid-state NMR suggested the cationic peptide predominated in the final assemblies. The cationic-to-anionic peptide ratio decreased with increasing charge. CATCH(2+) formed β-sheets when alone, whereas the other peptides remained unassembled. Fibrillization rate increased with peptide charge. The zwitterionic CATCH parent peptide, "Q11", assembled slowly and only at decreased simulation temperature. These results demonstrate that increasing charge draws complementary peptides together faster, favoring co-assembly, while like-charged molecules repel. We foresee these insights enabling development of co-assembled peptide biomaterials with defined content and predictable properties.
Collapse
Affiliation(s)
- Dillon T Seroski
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Xin Dong
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Kong M Wong
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Qing Shao
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Anant K Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Carol K Hall
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
31
|
Si Y, Tian Q, Zhao F, Kelly SH, Shores LS, Camacho DF, Sperling AI, Andrade MS, Collier JH, Chong AS. Adjuvant-free nanofiber vaccine induces in situ lung dendritic cell activation and T H17 responses. SCIENCE ADVANCES 2020; 6:eaba0995. [PMID: 32821819 PMCID: PMC7413739 DOI: 10.1126/sciadv.aba0995] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 06/25/2020] [Indexed: 05/07/2023]
Abstract
The current paradigm that subunit vaccines require adjuvants to optimally activate innate immunity implies that increased vaccine reactogenicity will invariably be linked to improved immunogenicity. Countering this paradigm, nanoparticulate vaccines have been reported to act as delivery systems for vaccine antigens and induce immunity without the need for exogenous adjuvants or local inflammation; however, the mechanisms underlying the immunogenicity of nanoparticle vaccines are incompletely identified. Here, we show that antigens displayed on self-assembling nanofiber scaffolds and delivered intranasally are presented by CD103+ and CD11b+ lung dendritic cells that up-regulate CD80 and migrate into the draining lymph node (LN). This was accompanied by a nearly exclusive priming and accumulation of antigen-specific TH17 cells occurring independently in both LN and lung. Thus, self-assembling peptide nanofiber vaccines may represent a novel, needle- and adjuvant-free means of eliciting protective immunity against fungal and bacterial infections at skin and mucosal barrier surfaces.
Collapse
Affiliation(s)
- Youhui Si
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiaomu Tian
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Fan Zhao
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Sean H. Kelly
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
| | - Lucas S. Shores
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
| | - Daniel F. Camacho
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Anne I. Sperling
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Michael S. Andrade
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Joel H. Collier
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
- Corresponding author. (A.S.C.); (J.H.C.)
| | - Anita S. Chong
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
- Corresponding author. (A.S.C.); (J.H.C.)
| |
Collapse
|
32
|
Biotin Functionalized Self‐Assembled Peptide Nanofiber as an Adjuvant for Immunomodulatory Response. Biotechnol J 2020; 15:e2000100. [DOI: 10.1002/biot.202000100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/15/2020] [Indexed: 12/20/2022]
|
33
|
Irvine DJ, Aung A, Silva M. Controlling timing and location in vaccines. Adv Drug Deliv Rev 2020; 158:91-115. [PMID: 32598970 PMCID: PMC7318960 DOI: 10.1016/j.addr.2020.06.019] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Vaccines are one of the most powerful technologies supporting public health. The adaptive immune response induced by immunization arises following appropriate activation and differentiation of T and B cells in lymph nodes. Among many parameters impacting the resulting immune response, the presence of antigen and inflammatory cues for an appropriate temporal duration within the lymph nodes, and further within appropriate subcompartments of the lymph nodes- the right timing and location- play a critical role in shaping cellular and humoral immunity. Here we review recent advances in our understanding of how vaccine kinetics and biodistribution impact adaptive immunity, and the underlying immunological mechanisms that govern these responses. We discuss emerging approaches to engineer these properties for future vaccines, with a focus on subunit vaccines.
Collapse
Affiliation(s)
- Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Murillo Silva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
34
|
Simonson AW, Aronson MR, Medina SH. Supramolecular Peptide Assemblies as Antimicrobial Scaffolds. Molecules 2020; 25:E2751. [PMID: 32545885 PMCID: PMC7355828 DOI: 10.3390/molecules25122751] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial discovery in the age of antibiotic resistance has demanded the prioritization of non-conventional therapies that act on new targets or employ novel mechanisms. Among these, supramolecular antimicrobial peptide assemblies have emerged as attractive therapeutic platforms, operating as both the bactericidal agent and delivery vector for combinatorial antibiotics. Leveraging their programmable inter- and intra-molecular interactions, peptides can be engineered to form higher ordered monolithic or co-assembled structures, including nano-fibers, -nets, and -tubes, where their unique bifunctionalities often emerge from the supramolecular state. Further advancements have included the formation of macroscopic hydrogels that act as bioresponsive, bactericidal materials. This systematic review covers recent advances in the development of supramolecular antimicrobial peptide technologies and discusses their potential impact on future drug discovery efforts.
Collapse
Affiliation(s)
- Andrew W. Simonson
- Department of Biomedical Engineering, The Pennsylvania State University, Suite 122, CBE Building, University Park, PA 16802-4400, USA; (A.W.S.); (M.R.A.)
| | - Matthew R. Aronson
- Department of Biomedical Engineering, The Pennsylvania State University, Suite 122, CBE Building, University Park, PA 16802-4400, USA; (A.W.S.); (M.R.A.)
| | - Scott H. Medina
- Department of Biomedical Engineering, The Pennsylvania State University, Suite 122, CBE Building, University Park, PA 16802-4400, USA; (A.W.S.); (M.R.A.)
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802-4400, USA
| |
Collapse
|
35
|
Huang Z, Song W, Chen X. Supramolecular Self-Assembled Nanostructures for Cancer Immunotherapy. Front Chem 2020; 8:380. [PMID: 32528926 PMCID: PMC7262496 DOI: 10.3389/fchem.2020.00380] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
Functional materials and nanostructures have been widely used for enhancing the therapeutic potency and safety of current cancer immunotherapy. While profound nanostructures have been developed to participate in the development of cancer immunotherapy, the construction of intricate nanostructures with easy fabrication and functionalization properties to satisfy the diversified requirements in cancer immunotherapy are highly required. Hierarchical self-assembly using supramolecular interactions to manufacture organized architectures at multiple length scales represents an interesting and promising avenue for sophisticated nanostructure construction. In this mini-review, we will outline the recent progress made in the development of supramolecular self-assembled nanostructures for cancer immunotherapy, with special focus on the supramolecular interactions including supramolecular peptide assembly, supramolecular DNA assembly, lipid hydrophobic assembly, host-guest assembly, and biomolecular recognition assembly.
Collapse
Affiliation(s)
- Zichao Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, China
| |
Collapse
|
36
|
Biomaterials for Immunoengineering. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00076-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
37
|
Okesola B, Wu Y, Derkus B, Gani S, Wu D, Knani D, Smith DK, Adams DJ, Mata A. Supramolecular Self-Assembly To Control Structural and Biological Properties of Multicomponent Hydrogels. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2019; 31:7883-7897. [PMID: 31631941 PMCID: PMC6792223 DOI: 10.1021/acs.chemmater.9b01882] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/08/2019] [Indexed: 05/07/2023]
Abstract
Self-assembled nanofibers are ubiquitous in nature and serve as inspiration for the design of supramolecular hydrogels. A multicomponent approach offers the possibility of enhancing the tunability and functionality of this class of materials. We report on the synergistic multicomponent self-assembly involving a peptide amphiphile (PA) and a 1,3:2,4-dibenzylidene-d-sorbitol (DBS) gelator to generate hydrogels with tunable nanoscale morphology, improved stiffness, enhanced self-healing, and stability to enzymatic degradation. Using induced circular dichroism of Thioflavin T (ThT), electron microscopy, small-angle neutron scattering, and molecular dynamics approaches, we confirm that the PA undergoes self-sorting, while the DBS gelator acts as an additive modifier for the PA nanofibers. The supramolecular interactions between the PA and DBS gelators result in improved bulk properties and cytocompatibility of the two-component hydrogels as compared to those of the single-component systems. The tunable mechanical properties, self-healing ability, resistance to proteolysis, and biocompatibility of the hydrogels suggest future opportunities for the hydrogels as scaffolds for tissue engineering and drug delivery vehicles.
Collapse
Affiliation(s)
- Babatunde
O. Okesola
- Institute
of Bioengineering, Queen Mary University
of London, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
| | - Yuanhao Wu
- Institute
of Bioengineering, Queen Mary University
of London, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
| | - Burak Derkus
- Institute
of Bioengineering, Queen Mary University
of London, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
- Biomedical
Engineering Department, Faculty of Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Samar Gani
- Department
of Biotechnology Engineering, ORT Braude
College, P.O. Box 78, Karmiel 2161002, Israel
| | - Dongsheng Wu
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
| | - Dafna Knani
- Department
of Biotechnology Engineering, ORT Braude
College, P.O. Box 78, Karmiel 2161002, Israel
| | - David K. Smith
- Department
of Chemistry, University of York, Heslington, York YO10 5DD, U.K.
| | - Dave J. Adams
- School
of
Chemistry, University of Glasgow, Glasgow G12 8QQ, U.K.
| | - Alvaro Mata
- Institute
of Bioengineering, Queen Mary University
of London, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary University of London, London E1 4NS, U.K.
| |
Collapse
|
38
|
Chen W, Yang S, Li S, Lang JC, Mao C, Kroll P, Tang L, Dong H. Self-Assembled Peptide Nanofibers Display Natural Antimicrobial Peptides to Selectively Kill Bacteria without Compromising Cytocompatibility. ACS APPLIED MATERIALS & INTERFACES 2019; 11:28681-28689. [PMID: 31328913 DOI: 10.1021/acsami.9b09583] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One of the major hurdles in the development of antimicrobial peptide (AMP)-based materials is their poor capacity in selectively killing bacteria without harming nearby mammalian cells. Namely, they are antimicrobial but cytotoxic. Current methods of nanoparticle-encapsulated AMPs to target bacteria selectively still have not yet overcome this hurdle. Here, we demonstrate a simple yet effective method to address this daunting challenge by associating a natural AMP with a β-sheet-forming synthetic peptide. The integrated peptides self-assembled to form a supramolecular nanofiber, resulting in the presentation of the AMP at the nanofiber-solvent interface in a precisely controlled manner. Using melittin as a model natural AMP, we found that the conformation of melittin changed dramatically when presented on the nanofiber surface, which, in turn, modulated the induced membrane permeability of the bacterial and mammalian cell membranes. Specifically, the presentation of melittin on the nanofiber restricted its hydrophobic residues, leading to a reduction of the hydrophobic interaction with lipids in the cell membranes. Compellingly, the reduced hydrophobic interaction led to a considerable decrease of melittin's induced permeability of the mammalian cell membrane than that of the bacterial cell membrane. As a result, the AMP-displaying nanofiber preferentially permeabilized and disrupted the membrane of the bacteria without compromising the mammalian cells. Such improved membrane selectivity and cytocompatibility were confirmed in a cell-based membrane localization and live-dead assay. Our new strategy holds great promise for fabricating cytocompatible antimicrobial assemblies that offer safer and more effective administration of therapeutic AMPs. These assemblies, with intrinsic antimicrobial activity and cytocompatibility, can also serve as building blocks for the construction of higher-ordered scaffolds for other biomedical applications such as tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
| | | | | | | | - Chuanbin Mao
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center , University of Oklahoma , Norman , Oklahoma 73019 , United States
| | | | | | | |
Collapse
|
39
|
Chakroun RW, Wang F, Lin R, Wang Y, Su H, Pompa D, Cui H. Fine-Tuning the Linear Release Rate of Paclitaxel-Bearing Supramolecular Filament Hydrogels through Molecular Engineering. ACS NANO 2019; 13:7780-7790. [PMID: 31117370 DOI: 10.1021/acsnano.9b01689] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
One key design feature in the development of any local drug delivery system is the controlled release of therapeutic agents over a certain period of time. In this context, we report the characteristic feature of a supramolecular filament hydrogel system that enables a linear and sustainable drug release over the period of several months. Through covalent linkage with a short peptide sequence, we are able to convert an anticancer drug, paclitaxel (PTX), to a class of prodrug hydrogelators with varying critical gelation concentrations. These self-assembling PTX prodrugs associate into filamentous nanostructures in aqueous conditions and consequently percolate into a supramolecular filament network in the presence of appropriate counterions. The intriguing linear drug release profile is rooted in the supramolecular nature of the self-assembling filaments which maintain a constant monomer concentration at the gelation conditions. We found that molecular engineering of the prodrug design, such as varying the number of oppositely charged amino acids or through the incorporation of hydrophobic segments, allows for the fine-tuning of the PTX linear release rate. In cell studies, these PTX prodrugs can exert effective cytotoxicity against glioblastoma cell lines and also primary brain cancer cells derived from patients and show enhanced tumor penetration in a cancer spheroid model. We believe this drug-bearing hydrogel platform offers an exciting opportunity for the local treatment of human diseases.
Collapse
Affiliation(s)
- Rami W Chakroun
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBiotechnology , The Johns Hopkins University , 3400 North Charles Street , Baltimore , Maryland 21218 , United States
| | - Feihu Wang
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBiotechnology , The Johns Hopkins University , 3400 North Charles Street , Baltimore , Maryland 21218 , United States
| | - Ran Lin
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBiotechnology , The Johns Hopkins University , 3400 North Charles Street , Baltimore , Maryland 21218 , United States
| | - Yin Wang
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBiotechnology , The Johns Hopkins University , 3400 North Charles Street , Baltimore , Maryland 21218 , United States
| | - Hao Su
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBiotechnology , The Johns Hopkins University , 3400 North Charles Street , Baltimore , Maryland 21218 , United States
| | - Danielle Pompa
- Department of Biomedical Engineering , University of Utah , 201 Presidents Circle , Salt Lake City , Utah 84112 , United States
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, and Institute for NanoBiotechnology , The Johns Hopkins University , 3400 North Charles Street , Baltimore , Maryland 21218 , United States
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center , Johns Hopkins University School of Medicine , Baltimore , Maryland 21205 , United States
- Center for Nanomedicine, The Wilmer Eye Institute , Johns Hopkins University School of Medicine , 400 North Broadway , Baltimore , Maryland 21231 , United States
| |
Collapse
|
40
|
Liu W, Wong-Noonan S, Pham NB, Pradhan I, Spigelmyer A, Funk R, Nedzesky J, Cohen H, Gawalt ES, Fan Y, Meng WS. A genetically engineered Fc-binding amphiphilic polypeptide for congregating antibodies in vivo. Acta Biomater 2019; 88:211-223. [PMID: 30822553 DOI: 10.1016/j.actbio.2019.02.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 01/08/2023]
Abstract
We report herein an affinity-based hydrogel used in creating subcutaneous depots of antibodies in vivo. The biomaterials design centered on pG_EAK, a polypeptide we designed and expressed in E. coli. The sequence consists of a truncated protein G (pG) genetically fused with repeats of the amphiphilic sequence AEAEAKAK ("EAK"). Capture of IgG was demonstrated in vitro in gels prepared from admixing pG_EAK and EAK ("pG_EAK/EAK gel"). The binding affinities and kinetics of pG for IgG were recapitulated in the pG_EAK polypeptide. Injecting IgG antibodies formulated with pG_EAK/EAK gel into subcutaneous space resulted in retention of the antibodies at the site for at least six days, whereas only signal at background levels was detected in grafts injected with IgG formulated in saline or diffusion-driven gel. The local retention of IgG in pG_EAK/EAK gel was correlated with limited distribution of the antibody in liver, spleen and lymph nodes, in contrast to those injected with antibodies formulated in saline or non-Fc binding EAK gel. In addition, antibodies formulated with pG_EAK/EAK gel and injected in mouse footpads were found to retain at the site for 19 days. As a demonstration of potential bioengineering applications, thymic epithelial cells (TECs), the primary population of thymic stromal cells that are critical for the development of T-lymphocytes, were mixed with pG_EAK/EAK gel formulated with TEC-specific anti-EpCAM antibodies and injected subcutaneously into athymic nude mice. The injected TECs congregated into functional thymic units in vivo, supporting the development of both CD4+ and CD8+ T cells as well as Foxp3+ regulatory T cells in the mice. In conclusion, pG_EAK/EAK gel can be used to retain IgG locally in vivo, and can be tailored as scaffolds for controlling deposition of molecular and/or cellular therapeutics. STATEMENT OF SIGNIFICANCE: The unique concept of the work centers on the genetic fusion of an Fc-binding domain and a self-assembling domain into a single polypeptide. To our knowledge, such bi-functional peptide has not been reported in the literature. The impact of the work lies in the ability to display IgG antibodies and Fc-fusion proteins of any specificity. The data shown demonstrate the platform can be used to localize IgG in vivo, and can be tailored for controlling deposition of primary thymic epithelial cells (TECs). The results support a biomaterials-based strategy by which TECs can be delivered as functional units to support T-lymphocyte development in vivo. The platform described in the study may serve as an important tool for immune engineering.
Collapse
|
41
|
Yang P, Song H, Feng Z, Wang C, Huang P, Zhang C, Kong D, Wang W. Synthetic, Supramolecular, and Self‐Adjuvanting CD8
+
T‐Cell Epitope Vaccine Increases the Therapeutic Antitumor Immunity. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Pengxiang Yang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical College Tianjin 300192 China
- Institute of Cancer Prevention and TreatmentHeilongjiang Academy of Medical ScienceHarbin Medical University Harbin 150081 China
| | - Huijuan Song
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical College Tianjin 300192 China
| | - Zujian Feng
- Department of Polymer Science and EngineeringCollege of Chemical Science and EngineeringTianjin University Tianjin 300072 China
| | - Changrong Wang
- Department of Polymer Science and EngineeringCollege of Chemical Science and EngineeringTianjin University Tianjin 300072 China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical College Tianjin 300192 China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical College Tianjin 300192 China
| | - Deling Kong
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical College Tianjin 300192 China
- State Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai University Tianjin 300071 China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical College Tianjin 300192 China
| |
Collapse
|
42
|
Morris C, Glennie SJ, Lam HS, Baum HE, Kandage D, Williams NA, Morgan DJ, Woolfson DN, Davidson AD. A Modular Vaccine Platform Combining Self-Assembled Peptide Cages and Immunogenic Peptides. ADVANCED FUNCTIONAL MATERIALS 2019; 29:1807357. [PMID: 32313545 PMCID: PMC7161841 DOI: 10.1002/adfm.201807357] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/18/2018] [Indexed: 05/11/2023]
Abstract
Subunit vaccines use delivery platforms to present minimal antigenic components for immunization. The benefits of such systems include multivalency, self-adjuvanting properties, and more specific immune responses. Previously, the design, synthesis, and characterization of self-assembling peptide cages (SAGEs) have been reported. In these, de novo peptides are combined to make hubs that assemble into nanoparticles when mixed in aqueous solution. Here it is shown that SAGEs are nontoxic particles with potential as accessible synthetic peptide scaffolds for the delivery of immunogenic components. To this end, SAGEs functionalized with the model antigenic peptides tetanus toxoid632-651 and ovalbumin323-339 drive antigen-specific responses both in vitro and in vivo, eliciting both CD4+ T cell and B cell responses. Additionally, SAGEs functionalized with the antigenic peptide hemagglutinin518-526 from the influenza virus are also able to drive a CD8+ T cell response in vivo. This work demonstrates the potential of SAGEs to act as a modular scaffold for antigen delivery, capable of inducing and boosting specific and tailored immune responses.
Collapse
Affiliation(s)
- Caroline Morris
- BrisSynBioUniversity of BristolBristolBS8 1TQUK
- School of ChemistryUniversity of BristolBristolBS8 1TSUK
| | - Sarah J. Glennie
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Hon S. Lam
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Holly E. Baum
- BrisSynBioUniversity of BristolBristolBS8 1TQUK
- School of ChemistryUniversity of BristolBristolBS8 1TSUK
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Dhinushi Kandage
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Neil A. Williams
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - David J. Morgan
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Derek N. Woolfson
- BrisSynBioUniversity of BristolBristolBS8 1TQUK
- School of ChemistryUniversity of BristolBristolBS8 1TSUK
- School of BiochemistryUniversity of BristolBristolBS8 1TDUK
| | - Andrew D. Davidson
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| |
Collapse
|
43
|
Lei K, Tang L. Surgery-free injectable macroscale biomaterials for local cancer immunotherapy. Biomater Sci 2019; 7:733-749. [DOI: 10.1039/c8bm01470a] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Up-to-date review and perspective on injectable macroscale biomaterials for local cancer immunotherapy.
Collapse
Affiliation(s)
- Kewen Lei
- Institute of Materials Science & Engineering
- École polytechnique fédérale de Lausanne (EPFL)
- Lausanne
- Switzerland
| | - Li Tang
- Institute of Materials Science & Engineering
- École polytechnique fédérale de Lausanne (EPFL)
- Lausanne
- Switzerland
- Institute of Bioengineering
| |
Collapse
|
44
|
Singha S, Shao K, Ellestad KK, Yang Y, Santamaria P. Nanoparticles for Immune Stimulation Against Infection, Cancer, and Autoimmunity. ACS NANO 2018; 12:10621-10635. [PMID: 30481968 DOI: 10.1021/acsnano.8b05950] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Vaccination using nanocarrier-based delivery systems has recently emerged as a promising approach for meeting the continued challenge posed by infectious diseases and cancer. A diverse portfolio of nanocarriers of various sizes, compositions, and physical parameters have now been developed, and this diversity provides an opportunity for the rational design of vaccines that can mediate targeted delivery of various antigens and adjuvants or immune regulatory agents in ways unachievable with classical vaccination approaches. This flexibility allows control over the characteristics of vaccine-elicited immune responses such that they can be tailored to be effective in circumstances where classical vaccines have failed. Furthermore, the utility of nanocarrier-based immune modulation extends to the treatment of autoimmune disease where precisely targeted inhibition of immune responses is desirable. Clearly, the selection of appropriate nanocarriers, antigens, adjuvants, and other components underpins the efficacy of these nanoimmune interventions. Herein, we provide an overview of currently available nanocarriers of various types and their physical and pharmacological properties with the goal of providing a resource for researchers exploring nanomaterial-based approaches for immune modulation and identify some information gaps and unexplored questions to help guide future investigation.
Collapse
Affiliation(s)
- Santiswarup Singha
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Kun Shao
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Kristofor K Ellestad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Yang Yang
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
- Institut D'Investigacions Biomèdiques August Pi i Sunyer , Barcelona 08036 , Spain
| |
Collapse
|
45
|
Zhou J, Yu G, Huang F. Supramolecular chemotherapy based on host-guest molecular recognition: a novel strategy in the battle against cancer with a bright future. Chem Soc Rev 2018; 46:7021-7053. [PMID: 28980674 DOI: 10.1039/c6cs00898d] [Citation(s) in RCA: 459] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chemotherapy is currently one of the most effective ways to treat cancer. However, traditional chemotherapy faces several obstacles to clinical trials, such as poor solubility/stability, non-targeting capability and uncontrollable release of the drugs, greatly limiting their anticancer efficacy and causing severe side effects towards normal tissues. Supramolecular chemotherapy integrating non-covalent interactions and traditional chemotherapy is a highly promising candidate in this regard and can be appropriately used for targeted drug delivery. By taking advantage of supramolecular chemistry, some limitations impeding traditional chemotherapy for clinical applications can be solved effectively. Therefore, we present here a review summarizing the progress of supramolecular chemotherapy in cancer treatment based on host-guest recognition and provide guidance on the design of new targeting supramolecular chemotherapy combining diagnostic and therapeutic functions. Based on a large number of state-of-the-art studies, our review will advance supramolecular chemotherapy on the basis of host-guest recognition and promote translational clinical applications.
Collapse
Affiliation(s)
- Jiong Zhou
- State Key Laboratory of Chemical Engineering, Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou 310027, P. R. China.
| | | | | |
Collapse
|
46
|
Al-Halifa S, Babych M, Zottig X, Archambault D, Bourgault S. Amyloid self-assembling peptides: Potential applications in nanovaccine engineering and biosensing. Pept Sci (Hoboken) 2018. [DOI: 10.1002/pep2.24095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Soultan Al-Halifa
- Department of Chemistry; Université du Québec à Montréal; Montreal, QC Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO; Québec QC Canada
| | - Margaryta Babych
- Department of Chemistry; Université du Québec à Montréal; Montreal, QC Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO; Québec QC Canada
| | - Ximena Zottig
- Department of Chemistry; Université du Québec à Montréal; Montreal, QC Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO; Québec QC Canada
| | - Denis Archambault
- Department of Biological Sciences; Université du Québec à Montréal; Montreal, QC Canada
- Swine and Poultry Infectious Diseases Research Centre, CRIPA; QC Canada
| | - Steve Bourgault
- Department of Chemistry; Université du Québec à Montréal; Montreal, QC Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO; Québec QC Canada
| |
Collapse
|
47
|
Kang SH, Hong SJ, Lee YK, Cho S. Oral Vaccine Delivery for Intestinal Immunity-Biological Basis, Barriers, Delivery System, and M Cell Targeting. Polymers (Basel) 2018; 10:E948. [PMID: 30960873 PMCID: PMC6403562 DOI: 10.3390/polym10090948] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/19/2022] Open
Abstract
Most currently available commercial vaccines are delivered by systemic injection. However, needle-free oral vaccine delivery is currently of great interest for several reasons, including the ability to elicit mucosal immune responses, ease of administration, and the relatively improved safety. This review summarizes the biological basis, various physiological and immunological barriers, current delivery systems with delivery criteria, and suggestions for strategies to enhance the delivery of oral vaccines. In oral vaccine delivery, basic requirements are the protection of antigens from the GI environment, targeting of M cells and activation of the innate immune response. Approaches to address these requirements aim to provide new vaccines and delivery systems that mimic the pathogen's properties, which are capable of eliciting a protective mucosal immune response and a systemic immune response and that make an impact on current oral vaccine development.
Collapse
Affiliation(s)
- Sung Hun Kang
- Department of Medical Sciences, College of Medicine, Hallym University, Chuncheon 24252, Korea.
| | - Seok Jin Hong
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong 18450, Korea.
| | - Yong-Kyu Lee
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea.
- 4D Biomaterials Center, Korea National University of Transportation, Jeungpyeong 27909, Korea.
| | - Sungpil Cho
- 4D Biomaterials Center, Korea National University of Transportation, Jeungpyeong 27909, Korea.
| |
Collapse
|
48
|
Zhang R, Kramer JS, Smith JD, Allen BN, Leeper CN, Li X, Morton LD, Gallazzi F, Ulery BD. Vaccine Adjuvant Incorporation Strategy Dictates Peptide Amphiphile Micelle Immunostimulatory Capacity. AAPS JOURNAL 2018; 20:73. [DOI: 10.1208/s12248-018-0233-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/14/2018] [Indexed: 12/26/2022]
|
49
|
Abstract
Self-assembled peptide nanostructures have been increasingly exploited as functional materials for applications in biomedicine and energy. The emergent properties of these nanomaterials determine the applications for which they can be exploited. It has recently been appreciated that nanomaterials composed of multicomponent coassembled peptides often display unique emergent properties that have the potential to dramatically expand the functional utility of peptide-based materials. This review presents recent efforts in the development of multicomponent peptide assemblies. The discussion includes multicomponent assemblies derived from short low molecular weight peptides, peptide amphiphiles, coiled coil peptides, collagen, and β-sheet peptides. The design, structure, emergent properties, and applications for these multicomponent assemblies are presented in order to illustrate the potential of these formulations as sophisticated next-generation bio-inspired materials.
Collapse
Affiliation(s)
- Danielle M Raymond
- Department of Chemistry, University of Rochester, Rochester, NY 14627-0216, USA.
| | | |
Collapse
|
50
|
Microwave-Assisted Synthesis and Immunological Evaluation of Self-Assembling Peptide Vaccines. Methods Mol Biol 2018. [PMID: 29744840 DOI: 10.1007/978-1-4939-7811-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Self-assembling peptides spontaneously associate into functional supramolecular scaffolds, which have found numerous biomedical applications. These molecular assemblies have applications in nerve regeneration, wound healing, and both prophylactic and therapeutic vaccination. They can also be useful tools for proliferation assays, sustained culture of difficult cell lines, or activation of cell lines for immunoassays. This protocol will describe the basic peptide synthesis and purification of model self-assembling peptide immunogen and methods for vaccinating mice, collecting lymph nodes, and stimulating cells ex vivo.
Collapse
|