1
|
Smith CT, Wang Z, Lewis JS. Engineering antigen-presenting cells for immunotherapy of autoimmunity. Adv Drug Deliv Rev 2024; 210:115329. [PMID: 38729265 DOI: 10.1016/j.addr.2024.115329] [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: 11/03/2023] [Revised: 03/05/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
Autoimmune diseases are burdensome conditions that affect a significant fraction of the global population. The hallmark of autoimmune disease is a host's immune system being licensed to attack its tissues based on specific antigens. There are no cures for autoimmune diseases. The current clinical standard for treating autoimmune diseases is the administration of immunosuppressants, which weaken the immune system and reduce auto-inflammatory responses. However, people living with autoimmune diseases are subject to toxicity, fail to mount a sufficient immune response to protect against pathogens, and are more likely to develop infections. Therefore, there is a concerted effort to develop more effective means of targeting immunomodulatory therapies to antigen-presenting cells, which are involved in modulating the immune responses to specific antigens. In this review, we highlight approaches that are currently in development to target antigen-presenting cells and improve therapeutic outcomes in autoimmune diseases.
Collapse
Affiliation(s)
- Clinton T Smith
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Zhenyu Wang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Jamal S Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| |
Collapse
|
2
|
Firdessa-Fite R, Johnson SN, Bechi Genzano C, Leon MA, Ku A, Ocampo Gonzalez FA, Milner JD, Sestak JO, Berkland C, Creusot RJ. Soluble antigen arrays provide increased efficacy and safety over free peptides for tolerogenic immunotherapy. Front Immunol 2024; 15:1258369. [PMID: 38933266 PMCID: PMC11199391 DOI: 10.3389/fimmu.2024.1258369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Autoantigen-specific immunotherapy using peptides offers a more targeted approach to treat autoimmune diseases, but clinical implementation has been challenging. We previously showed that multivalent delivery of peptides as soluble antigen arrays (SAgAs) efficiently protects against spontaneous autoimmune diabetes in the non-obese diabetic (NOD) mouse model. Here, we compared the efficacy, safety, and mechanisms of action of SAgAs versus free peptides. SAgAs, but not their corresponding free peptides at equivalent doses, efficiently prevented the development of diabetes. SAgAs increased the frequency of regulatory T cells among peptide-specific T cells or induce their anergy/exhaustion or deletion, depending on the type of SAgA used (hydrolysable (hSAgA) and non-hydrolysable 'click' SAgA (cSAgA)) and duration of treatment, whereas their corresponding free peptides induced a more effector phenotype following delayed clonal expansion. Over time, the peptides induced an IgE-independent anaphylactic reaction, the incidence of which was significantly delayed when peptides were in SAgA form rather than in free form. Moreover, the N-terminal modification of peptides with aminooxy or alkyne linkers, which was needed for grafting onto hyaluronic acid to make hSAgA or cSAgA variants, respectively, influenced their stimulatory potency and safety, with alkyne-functionalized peptides being more potent and less anaphylactogenic than aminooxy-functionalized peptides. Immunologic anaphylaxis occurred in NOD mice in a dose-dependent manner but not in C57BL/6 or BALB/c mice; however, its incidence did not correlate with the level of anti-peptide antibodies. We provide evidence that SAgAs significantly improve the efficacy of peptides to induce tolerance and prevent autoimmune diabetes while at the same time reducing their anaphylactogenic potential.
Collapse
Affiliation(s)
- Rebuma Firdessa-Fite
- Columbia Center for Translational Immunology, Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Stephanie N. Johnson
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Camillo Bechi Genzano
- Columbia Center for Translational Immunology, Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Martin A. Leon
- Department of Chemistry, University of Kansas, Lawrence, KS, United States
| | - Amy Ku
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and New York Presbyterian Hospital, New York, NY, United States
| | - Fernando A. Ocampo Gonzalez
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and New York Presbyterian Hospital, New York, NY, United States
| | - Joshua D. Milner
- Department of Pediatrics, Division of Allergy and Immunology, Columbia University Irving Medical Center, New York, NY, United States
| | - Joshua O. Sestak
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, United States
| | - Remi J. Creusot
- Columbia Center for Translational Immunology, Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
3
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [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: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
4
|
Wemlinger SM, Cambier JC. Therapeutic tactics for targeting B lymphocytes in autoimmunity and cancer. Eur J Immunol 2024; 54:e2249947. [PMID: 37816494 DOI: 10.1002/eji.202249947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/12/2023]
Abstract
B lymphocytes have become a very popular therapeutic target in a number of autoimmune indications due to their newly appreciated roles, and approachability, in these diseases. Many of the therapies now applied in autoimmunity were initially developed to deplete malignant B cells. These strategies have also been found to benefit patients suffering from such autoimmune diseases as multiple sclerosis, type I diabetes, systemic lupus erythematosus, and rheumatoid arthritis, to name a few. These observations have supported the expansion of research addressing the mechanistic contributions of B cells in these diseases, as well as blossoming of therapeutics that target them. This review seeks to summarize cutting-edge modalities for targeting B cells, including monoclonal antibodies, bispecific antibodies, antibody-drug conjugates, chimeric antigen receptor-T cells, and small molecule inhibitors. Efforts to refine B-cell targeted therapy to eliminate only pathogenic autoreactive cells will be addressed as well as the potential for future B-cell-based cellular therapeutics. Finally, we also address approaches that seek to silence B-cell function without depletion.
Collapse
Affiliation(s)
- Scott M Wemlinger
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
5
|
Johnson SN, Brucks SD, Apley KD, Farrell MP, Berkland CJ. Multivalent Scaffolds to Promote B cell Tolerance. Mol Pharm 2023; 20:3741-3756. [PMID: 37410969 DOI: 10.1021/acs.molpharmaceut.3c00039] [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: 07/08/2023]
Abstract
Autoimmune diseases are characterized by aberrant immune responses toward self-antigens. Current treatments lack specificity, promoting adverse effects by broadly suppressing the immune system. Therapies that specifically target the immune cells responsible for disease are a compelling strategy to mitigate adverse effects. Multivalent formats that display numerous binding epitopes off a single scaffold may enable selective immunomodulation by eliciting signals through pathways unique to the targeted immune cells. However, the architecture of multivalent immunotherapies can vary widely, and there is limited clinical data with which to evaluate their efficacy. Here, we set forth to review the architectural properties and functional mechanisms afforded by multivalent ligands and evaluate four multivalent scaffolds that address autoimmunity by altering B cell signaling pathways. First, we address both synthetic and natural polymer backbones functionalized with a variety of small molecule, peptide, and protein ligands for probing the effects of valency and costimulation. Then, we review nanoparticles composed entirely from immune signals which have been shown to be efficacious. Lastly, we outline multivalent liposomal nanoparticles capable of displaying high numbers of protein antigens. Taken together, these examples highlight the versatility and desirability of multivalent ligands for immunomodulation and illuminate strengths and weaknesses of multivalent scaffolds for treating autoimmunity.
Collapse
Affiliation(s)
- Stephanie N Johnson
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Spencer D Brucks
- Department of Chemistry, Harvey Mudd College, Claremont, California 91711, United States
| | - Kyle D Apley
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Mark P Farrell
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
| | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047, United States
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
- Bioengineering Program, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
6
|
Holborough-Kerkvliet MD, Kroos S, de Wetering RV, Toes REM. Addressing the key issue: Antigen-specific targeting of B cells in autoimmune diseases. Immunol Lett 2023:S0165-2478(23)00075-5. [PMID: 37209914 DOI: 10.1016/j.imlet.2023.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Autoimmune diseases are heterogeneous pathologies characterized by a breakdown of immunological tolerance to self, resulting in a chronic and aberrant immune response to self-antigens. The scope and extent of affected tissues can vary greatly per autoimmune disease and can involve multiple organs and tissue types. The pathogenesis of most autoimmune diseases remains unknown but it is widely accepted that a complex interplay between (autoreactive) B and T cells in the context of breached immunological tolerance drives autoimmune pathology. The importance of B cells in autoimmune disease is exemplified by the successful use of B cell targeting therapies in the clinic. For example, Rituximab, a depleting anti-CD20 antibody, has shown favorable results in reducing the signs and symptoms of multiple autoimmune diseases, including Rheumatoid Arthritis, Anti-Neutrophil Cytoplasmic Antibody associated vasculitis and Multiple Sclerosis. However, Rituximab depletes the entire B cell repertoire, leaving patients susceptible to (latent) infections. Therefore, multiple ways to target autoreactive cells in an antigen-specific manner are currently under investigation. In this review, we will lay out the current state of antigen-specific B cell inhibiting or depleting therapies in the context of autoimmune diseases.
Collapse
Affiliation(s)
| | - Sanne Kroos
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Renee van de Wetering
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
7
|
Firdessa-Fite R, Johnson SN, Leon MA, Sestak JO, Berkland C, Creusot RJ. Soluble antigen arrays improve the efficacy and safety of peptide-based tolerogenic immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539161. [PMID: 37205572 PMCID: PMC10187310 DOI: 10.1101/2023.05.05.539161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Autoantigen-specific immunotherapy using peptides offers a more targeted approach to treat autoimmune diseases, but the limited in vivo stability and uptake of peptides impedes clinical implementation. We previously showed that multivalent delivery of peptides as soluble antigen arrays (SAgAs) efficiently protects against spontaneous autoimmune diabetes in the non-obese diabetic (NOD) mouse model. Here, we compared the efficacy, safety, and mechanisms of action of SAgAs versus free peptides. SAgAs, but not their corresponding free peptides at equivalent doses, efficiently prevented the development of diabetes. SAgAs increased the frequency of regulatory T cells among peptide-specific T cells or induce their anergy/exhaustion or deletion, depending on the type of SAgA (hydrolysable (hSAgA) and non-hydrolysable 'click' SAgA (cSAgA)) and duration of treatment, whereas their corresponding free peptides induced a more effector phenotype following delayed clonal expansion. Moreover, the N-terminal modification of peptides with aminooxy or alkyne linkers, which was needed for grafting onto hyaluronic acid to make hSAgA or cSAgA variants, respectively, influenced their stimulatory potency and safety, with alkyne-functionalized peptides being more potent and less anaphylactogenic than aminooxy-functionalized peptides. Both SAgA variants significantly delayed anaphylaxis compared to their respective free peptides. The anaphylaxis, which occurred in NOD mice but not in C57BL/6 mice, was dose-dependent but did not correlate with the production of IgG1 or IgE against the peptides. We provide evidence that SAgAs significantly improve the efficacy and safety of peptide-based immunotherapy.
Collapse
Affiliation(s)
- Rebuma Firdessa-Fite
- Columbia Center for Translational Immunology, Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Medical Center, 650 West 168 St, New York, NY 10032
| | - Stephanie N. Johnson
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047
| | - Martin A. Leon
- Department of Chemistry, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS 66045
| | - Joshua O. Sestak
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047
- Department of Chemical and Petroleum Engineering, University of Kansas,1530 West 15 Street, Lawrence, KS 66045
| | - Remi J. Creusot
- Columbia Center for Translational Immunology, Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Medical Center, 650 West 168 St, New York, NY 10032
| |
Collapse
|
8
|
Wu Y, Huang J, He H, Wang M, Yin G, Qi L, He X, Wang HH, Wang K. Logic Nanodevice-Mediated Receptor Assembly for Nongenetic Regulation of Cell Behavior in Tumor-like Microenvironment. NANO LETTERS 2023; 23:1801-1809. [PMID: 36826373 DOI: 10.1021/acs.nanolett.2c04657] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The reprogramming of cell signaling and behavior through the artificial control of cell surface receptor oligomerization shows great promise in biomedical research and cell-based therapy. However, it remains challenging to achieve combinatorial recognition in a complicated environment and logical regulation of receptors for desirable cellular behavior. Herein, we develop a logic-gated DNA nanodevice with responsiveness to multiple environmental inputs for logically controlled assembly of heterogeneous receptors to modulate signaling. The "AND" gate nanodevice uses an i-motif and an ATP-binding aptamer as environmental cue-responsive units, which can successfully implement a logic operation to manipulate receptors on the cell surface. In the presence of both protons and ATP, the DNA nanodevice is activated to selectively assemble MET and CD71, which modulate the HGF/MET signaling, resulting in cytoskeletal reorganization to inhibit cancer cell motility in a tumor-like microenvironment. Our strategy would be highly promising for precision therapeutics, including controlled drug release and cancer treatment.
Collapse
Affiliation(s)
- Yuchen Wu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| | - Jin Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| | - Hui He
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| | - Meixia Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| | - Guanyu Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| | - Lanlin Qi
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| | - Xiaoxiao He
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| | - Hong-Hui Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| | - Kemin Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Key Laboratory for Bio-Nanotechnology and Molecule Engineering of Hunan Province, Changsha 410082, China
| |
Collapse
|
9
|
Zhang S, Ouyang T, Reinhard BM. Multivalent Ligand-Nanoparticle Conjugates Amplify Reactive Oxygen Species Second Messenger Generation and Enhance Epidermal Growth Factor Receptor Phosphorylation. Bioconjug Chem 2022; 33:1716-1728. [PMID: 35993676 PMCID: PMC9815836 DOI: 10.1021/acs.bioconjchem.2c00335] [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] [Indexed: 01/11/2023]
Abstract
The epidermal growth factor (EGF) receptor (EGFR) is heterogeneously distributed on the cellular surface and enriched in clusters with diameters of tens of nanometers. Multivalent presentation of EGF ligand on nanoparticles (NPs) provides an approach for controlling and amplifying the local activation of EGFR in these clusters. Reactive oxygen species (ROS) have been indicated to play a role in the regulation of EGFR activation as second messengers, but the effect of nanoconjugation on EGF-mediated ROS formation and ROS-induced EGFR activation is not well established. The goal of this manuscript is to characterize the multivalent enhancement of EGF-induced ROS formation and to test its effect on EGFR phosphorylation in breast cancer cell models using gold (Au) NPs with a diameter of 81 ± 1 nm functionalized with two different EGF ligand densities (12 ± 7 EGF/NP (NP-EGF12) and 87 ± 6 EGF/NP (NP-EGF87)). In the EGFR overexpressing cell lines MDA-MB-231 and MDA-MB-468, NP-EGF87 achieved a measurable multivalent enhancement of ROS that peaked at concentrations c ROSmax ≤ 25 pM and that were EGFR and nicotinamide adenine dinucleotide phosphate oxidase (NOX) dependent. NP-EGF12 failed to generate comparable ROS levels as NP-EGF87 in the investigated NP input concentration range (0-100 pM). In cells with nearly identical numbers of bound NP-EGF87 and NP-EGF12, the ROS levels for NP-EGF87 were systematically higher, indicating that the multivalent enhancement is exclusively related not only to avidity but also to a stronger stimulation per NP. Importantly, the increase in EGF-induced ROS formation associated with EGF nanoconjugation at c ROSmax resulted in a measurable gain in EGFR phosphorylation, confirming that ROS generation contributes to the multivalent enhancement of EGFR activation in response to NP-EGF87.
Collapse
Affiliation(s)
- Sandy Zhang
- Department of Chemistry and The Photonics Center, Boston University, Boston, MA 02215
| | - Tianhong Ouyang
- Department of Chemistry and The Photonics Center, Boston University, Boston, MA 02215
| | - Björn M. Reinhard
- Department of Chemistry and The Photonics Center, Boston University, Boston, MA 02215
| |
Collapse
|
10
|
He Q, Liu Y, Li K, Wu Y, Wang T, Tan Y, Jiang T, Liu X, Liu Z. Deoxyribonucleic acid anchored on cell membranes for biomedical application. Biomater Sci 2021; 9:6691-6717. [PMID: 34494042 DOI: 10.1039/d1bm01057c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Engineering cellular membranes with functional molecules provides an attractive strategy to manipulate cellular behaviors and functionalities. Currently, synthetic deoxyribonucleic acid (DNA) has emerged as a promising molecular tool to engineer cellular membranes for biomedical applications due to its molecular recognition and programmable properties. In this review, we summarized the recent advances in anchoring DNA on the cellular membranes and their applications. The strategies for anchoring DNA on cell membranes were summarized. Then their applications, such as immune response activation, receptor oligomerization regulation, membrane structure mimicking, cell-surface biosensing, and construction of cell clusters, were listed. The DNA-enabled intelligent systems which were able to sense stimuli such as DNA strands, light, and metal ions were highlighted. Finally, insights regarding the remaining challenges and possible future directions were provided.
Collapse
Affiliation(s)
- Qunye He
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Ke Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yuwei Wu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Ting Wang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yifu Tan
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Ting Jiang
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Xiaoqin Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China. .,Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, P. R. China
| |
Collapse
|
11
|
Therapeutic Targeting of Autoreactive B Cells: Why, How, and When? Biomedicines 2021; 9:biomedicines9010083. [PMID: 33467130 PMCID: PMC7829839 DOI: 10.3390/biomedicines9010083] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 11/20/2022] Open
Abstract
B lymphocytes play critical roles in the development of autoimmunity, acting as autoantibody manufacturers, antigen-presenting cells, and producers of cytokines. Pan-B cell depletion has demonstrated efficacy in treatment of many autoimmune disorders, but carries with it an unfavorable safety profile due to global immune suppression. Hence, attention has turned to the potential of autoantigen-specific B cell targeted therapies, which would deplete or silence pathogenic self-antigen-reactive cells while sparing B cells needed for immune defense. Here, we discuss the antigen-specific B cell-targeted approaches that are under development or are under consideration, that could be employed to allow for more precise therapy in the treatment of autoimmunity. Lastly, we discuss some of the challenges associated with antigen-specific B cell targeting that may impact their clinical applicability.
Collapse
|
12
|
Christopher MA, Johnson SN, Griffin JD, Berkland CJ. Autoantigen Tetramer Silences Autoreactive B Cell Populations. Mol Pharm 2020; 17:4201-4211. [PMID: 32903002 DOI: 10.1021/acs.molpharmaceut.0c00665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Many autoimmune therapies focus on immune suppression to reduce symptom severity and halt disease progression; however, currently approved treatments lack specificity for the autoantigen and rely on more global immune suppression. Multivalent antigen arrays can disarm pathogenic autoimmune B cell populations that specifically recognize the antigen of interest via their B cell receptor (BCR). Disarmament may be achieved by BCR engagement, cross-linking, and sustained receptor occupancy as a result of multivalent, high avidity BCR binding. To engage and explore this mechanism, a tetramer display of the encephalogenic proteolipid peptide (PLP139-151), referred to as 4-arm PLP139-151, was synthesized by copper-catalyzed azide-alkyne cycloaddition chemistry. Subcutaneous administration of 4-arm PLP139-151 completely ameliorated symptoms of paralysis in a mouse model of multiple sclerosis known as experimental autoimmune encephalomyelitis. Competitive binding of 4-arm PLP139-151 to PLP139-151-specific IgG in the mouse serum demonstrated the enhanced avidity associated with the multivalent array compared to the free peptide. Furthermore, key PLP139-151-reactive B cells were depleted following 4-arm PLP139-151 treatment, resulting in significant reduction of proinflammatory cytokines. Together, these data demonstrate the potential of 4-arm PLP139-151 to silence autoreactive B cell populations and limit the downstream activation of effector cells.
Collapse
Affiliation(s)
- Matthew A Christopher
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| | - Stephanie N Johnson
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| | - J Daniel Griffin
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States.,Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, Kansas 66045, United States
| | - Cory J Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States.,Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, Kansas 66045, United States.,Chemical and Petroleum Engineering, University of Kansas, 1520 West 15th Street, Lawrence, Kansas 66045, United States
| |
Collapse
|
13
|
Johnson SN, Griffin JD, Hulbert C, DeKosky BJ, Thomas JW, Berkland CJ. Multimeric Insulin Desensitizes Insulin-Specific B Cells. ACS APPLIED BIO MATERIALS 2020; 3:6319-6330. [DOI: 10.1021/acsabm.0c00782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Stephanie N. Johnson
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - J. Daniel Griffin
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas 66045, United States
| | - Chrys Hulbert
- Department of Medicine, Division of Rheumatology, and Immunology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Brandon J. DeKosky
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| | - James W. Thomas
- Department of Medicine, Division of Rheumatology, and Immunology, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Cory J. Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
14
|
Griffin JD, Song JY, Sestak JO, DeKosky BJ, Berkland CJ. Linking autoantigen properties to mechanisms of immunity. Adv Drug Deliv Rev 2020; 165-166:105-116. [PMID: 32325104 PMCID: PMC7572523 DOI: 10.1016/j.addr.2020.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023]
Abstract
Antigen-specific immunotherapies (ASIT) present compelling potential for introducing precision to the treatment of autoimmune diseases where nonspecific, global immunosuppression is currently the only treatment option. Central to ASIT design is the delivery of autoantigen, which parallels allergy desensitization approaches. Clinical success in tolerizing allergen-specific responses spans longer than a century, but autoimmune ASITs have yet to see an FDA-approved breakthrough. Allergens and autoantigens differ substantially in physicochemical properties, and these discrepancies influence the nature of their interactions with the immune system. Approved allergen-specific immunotherapies are typically administered as water soluble, neutrally charged protein fractions from 10 to 70 kDa. Conversely, autoantigens are native proteins that exhibit wide-ranging sizes, solubilities, and charges that render them susceptible to immunogenicity. To translate the success of allergen hyposensitization to ASIT, delivery strategies may be necessary to effectively format autoantigens, guide biodistribution, and engage appropriate immune mechanisms.
Collapse
Affiliation(s)
- J Daniel Griffin
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, United States of America; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America
| | - Jimmy Y Song
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America; Bioengineering Graduate Program, University of Kansas, Lawrence, KS, United States of America
| | - Joshua O Sestak
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America; Orion BioScience, Inc, Omaha, NE, United States of America
| | - Brandon J DeKosky
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, United States of America; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, United States of America
| | - Cory J Berkland
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, United States of America; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States of America; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, United States of America.
| |
Collapse
|
15
|
Li J, Wang L, Tian J, Zhou Z, Li J, Yang H. Nongenetic engineering strategies for regulating receptor oligomerization in living cells. Chem Soc Rev 2020; 49:1545-1568. [DOI: 10.1039/c9cs00473d] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nongenetic strategies for regulating receptor oligomerization in living cells based on DNA, protein, small molecules and physical stimuli.
Collapse
Affiliation(s)
- Jingying Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Liping Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Jinmiao Tian
- Institute of Molecular Medicine
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
- Shanghai
| | - Zhilan Zhou
- Institute of Molecular Medicine
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
- Shanghai
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology
- Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety
- State Key Laboratory of Photocatalysis on Energy and Environment
- College of Chemistry
- Fuzhou University
| |
Collapse
|
16
|
Griffin JD, Song JY, Huang A, Sedlacek AR, Flannagan KL, Berkland CJ. Antigen-specific immune decoys intercept and exhaust autoimmunity to prevent disease. Biomaterials 2019; 222:119440. [PMID: 31450159 DOI: 10.1016/j.biomaterials.2019.119440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/31/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022]
Abstract
Relapsing-remitting patterns of many autoimmune diseases such as multiple sclerosis (MS) are perpetuated by a recurring circuit of adaptive immune cells that amplify in secondary lymphoid organs (SLOs) and traffic to compartments where antigen is abundant to elicit damage. Some of the most effective immunotherapies impede the migration of immune cells through this circuit, however, broadly suppressing immune cell migration can introduce life-threatening risks for patients. We developed antigen-specific immune decoys (ASIDs) to mimic tissues targeted in autoimmunity and selectively intercept autoimmune cells to preserve host tissue. Using Experimental Autoimmune Encephalomyelitis (EAE) as a model, we conjugated autoantigen PLP139-151 to a microporous collagen scaffold. By subcutaneously implanting ASIDs after induction but prior to the onset of symptoms, mice were protected from paralysis. ASID implants were rich with autoimmune cells, however, reactivity to cognate antigen was substantially diminished and apoptosis was prevalent. ASID-implanted mice consistently exhibited engorged spleens when disease normally peaked. In addition, splenocyte antigen-presenting cells were highly activated in response to PLP rechallenge, but CD3+ and CD19 + effector subsets were significantly decreased, suggesting exhaustion. ASID-implanted mice never developed EAE relapse symptoms even though the ASID material had long since degraded, suggesting exhausted autoimmune cells did not recover functionality. Together, data suggested ASIDs were able to sequester and exhaust immune cells in an antigen-specific fashion, thus offering a compelling approach to inhibit the migration circuit underlying autoimmunity.
Collapse
Affiliation(s)
- J Daniel Griffin
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA
| | - Jimmy Y Song
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Aric Huang
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Alexander R Sedlacek
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, USA
| | - Kaitlin L Flannagan
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, USA
| | - Cory J Berkland
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA; Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
17
|
Leon MA, Firdessa-Fite R, Ruffalo JK, Pickens CJ, Sestak JO, Creusot RJ, Berkland C. Soluble Antigen Arrays Displaying Mimotopes Direct the Response of Diabetogenic T Cells. ACS Chem Biol 2019; 14:1436-1448. [PMID: 31260253 DOI: 10.1021/acschembio.9b00090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder which develops when insulin-producing, pancreatic beta cells are destroyed by an aberrant immune response. Current therapies for T1D either treat symptoms or cause global immunosuppression, which leave patients at risk of developing long-term complications or vulnerable to foreign pathogens. Antigen-specific immunotherapies have emerged as a selective approach for autoimmune diseases by inducing tolerance while mitigating global immunosuppression. We previously reported SAgAs with multiple copies of a multiple sclerosis (MS) autoantigen grafted onto hyaluronic acid (HA) as an efficacious therapy in experimental autoimmune encephalomyelitis. While the immune response of MS is distinct from that of T1D, the mechanism of SAgAs was hypothesized to be similar and via induction of immune tolerance to diabetes antigens. We synthesized SAgAs composed of HA polymer backbone conjugated with multiple copies of the T1D autoantigen mimotope p79 using aminooxy chemistry (SAgAp79) or using copper-catalyzed alkyne-azide cycloaddition (cSAgAp79) chemistry. SAgAs constructed using the hydrolyzable aminooxy linkage, thus capable of releasing p79, exhibited physicochemical properties similar to the triazole linkage. Both SAgAp79 versions showed high specificity and efficacy in stimulating epitope-specific T cells. SAgAs can be taken up by most immune cell populations but do not induce their maturation, and conventional dendritic cells are responsible for the brunt of antigen presentation within splenocytes. cSAgAp79 was more stimulatory than SAgAp79 both in vitro and in vivo, an effect that was ascribed to the peptide modification rather than the type of linkage. In summary, we provide here the first proof-of-principle that SAgA therapy could also be applicable to T1D.
Collapse
Affiliation(s)
- Martin A. Leon
- Department of Chemistry, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| | - Rebuma Firdessa-Fite
- Columbia Center for Translational Immunology, Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Medical Center, 650 West 168th Street, New York, New York 10032, United States
| | - Justin K. Ruffalo
- Department of Chemical and Petroleum Engineering, University of Kansas, 1530 West 15th Street, Lawrence, Kansas 66045, United States
| | - Chad J. Pickens
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| | - Joshua O. Sestak
- Orion BioScience Inc., 986099 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Remi J. Creusot
- Columbia Center for Translational Immunology, Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Medical Center, 650 West 168th Street, New York, New York 10032, United States
| | - Cory Berkland
- Department of Chemistry, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, Kansas 66047, United States
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, Kansas 66045, United States
- Department of Chemical and Petroleum Engineering, University of Kansas, 1530 West 15th Street, Lawrence, Kansas 66045, United States
| |
Collapse
|
18
|
Griffin JD, Leon MA, Salash JR, Shao M, Hartwell BL, Pickens CJ, Sestak JO, Berkland C. Acute B-Cell Inhibition by Soluble Antigen Arrays Is Valency-Dependent and Predicts Immunomodulation in Splenocytes. Biomacromolecules 2019; 20:2115-2122. [PMID: 30995843 DOI: 10.1021/acs.biomac.9b00328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Antigen valency plays a fundamental role in directing the nature of an immune response to be stimulatory or tolerogenic. Soluble antigen arrays (SAgAs) are an antigen-specific immunotherapy that combats autoimmunity through the multivalent display of autoantigen. Although mechanistic studies have shown SAgAs to induce T- and B-cell anergy, the effect of SAgA valency has never been experimentally tested. Here, SAgAs of discrete antigen valencies were synthesized by click chemistry and evaluated for acute B-cell signaling inhibition as well as downstream immunomodulatory effects in splenocytes. Initial studies using the Raji B-cell line demonstrated SAgA valency dictated the extent of calcium flux. Lower valency constructs elicited the largest reductions in B-cell activation. In splenocytes from mice with experimental autoimmune encephalomyelitis, the same valency-dependent effects were evident in the downregulation of the costimulatory marker CD86. The reduction of calcium flux observed in Raji B-cells correlated strongly with downregulation in splenocyte CD86 expression after 72 h. Here, a thorough analysis of SAgA antigenic valency illustrates that low, but not monovalent, presentation of autoantigen was ideal for eliciting the most potent immunomodulatory effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Cory Berkland
- Orion BioScience , Omaha , Nebraska 68198 , United States
| |
Collapse
|
19
|
Zhang Y, Cheng M, Cao J, Zhang Y, Yuan Z, Wu Q, Wang W. Multivalent nanoparticles for personalized theranostics based on tumor receptor distribution behavior. NANOSCALE 2019; 11:5005-5013. [PMID: 30839969 DOI: 10.1039/c8nr09347d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
It is acknowledged that the targeting ability of multivalent ligand-modified nanoparticles (MLNs) strongly depends on the ligand spatial presentation determined by ligand valency. However, the receptor overexpression level varies between different types or stages of tumors. Thus, it is essential to explore the influence of ligand valency on the targeting ability of MLNs to tumors with different levels of receptor overexpression. In this study, a dual-acting agent raltitrexed was used as a ligand to target the folate receptor (FR). Different copies of the raltitrexed-modified multivalent dendritic polyethyleneimine ligand cluster PRn (n = 2, 4, and 8) were conjugated onto magnetic nanoparticles to form multivalent magnetic NPs (MMNs) with different valences. The in vitro studies demonstrated that Fe-PR4 was the most effective valency in the treatment of high FR overexpressing KB cells with a decentralized receptor distribution, owing to the fact that Fe-PR2 was negative in statistical rebinding and Fe-PR8 could induce steric hindrance in the limited binding area. Instead, in moderate FR overexpressing HeLa cells with clustered receptor display, the extra ligands on Fe-PR8 would facilitate statistical rebinding more beneficially. Furthermore, in in vivo tumor inhibition and targeted magnetic resonance imaging (MRI) of KB tumors and another moderate FR expressing H22 tumor, similar results were obtained with the cell experiments. Overall, the optimizable treatment effect of Fe-PRn by modulating the ligand valency based on the overexpressing tumor receptor distribution behavior supports the potential of Fe-PRn as a nanomedicine for personalized theranostics.
Collapse
Affiliation(s)
- Yahui Zhang
- Key Laboratory of Functional Polymer Materials of the Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | | | | | | | | | | | | |
Collapse
|
20
|
Leon MA, Wemlinger SM, Larson NR, Ruffalo JK, Sestak JO, Middaugh CR, Cambier JC, Berkland C. Soluble Antigen Arrays for Selective Desensitization of Insulin-Reactive B Cells. Mol Pharm 2019; 16:1563-1572. [PMID: 30681867 DOI: 10.1021/acs.molpharmaceut.8b01250] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Autoimmune diseases are believed to be highly dependent on loss of immune tolerance to self-antigens. Currently, no treatments have been successful clinically in inducing autoantigen-specific tolerance, including efforts to utilize antigen-specific immunotherapy (ASIT) to selectively correct the aberrant autoimmunity. Soluble antigen arrays (SAgAs) represent a novel autoantigen delivery system composed of a linear polymer, hyaluronic acid (HA), displaying multiple copies of conjugated autoantigen. We have previously reported that soluble antigen arrays displaying proteolipid peptide (SAgAPLP) induced tolerance to this specific multiple sclerosis (MS) autoantigen. Utilizing SAgA technology, we have developed a new ASIT as a possible type 1 diabetes (T1D) therapeutic by conjugating human insulin to HA, known as soluble antigen array insulin (SAgAIns). Three types were synthesized, low valency lvSAgAIns (2 insulins per HA), medium valency mvSAgAIns (4 insulins per HA), and, high valency hvSAgAIns (9 insulins per HA), to determine if valency differentially modulates the ex vivo activity of insulin-binding B cells (IBCs). Extensive biophysical characterization was performed for the SAgA molecules. SAgAIns molecules were successfully used to affect the biologic activity of IBCs by inducing desensitization of the B cell antigen receptors (BCR). SAgAIns bound specifically to insulin-reactive B cells without blocking epitopes recognized by antibodies against the Fc regions of membrane immunoglobulin or CD79 transducer components of the BCR. Preincubation of IBCs (125Tg) with SAgAIns, but not HA alone, rendered the IBCs refractory to restimulation. SAgAIns induced a decrease in BCR expression and IP3R-mediated intracellular calcium release. Surprisingly, SAgAIns binding to BCR on the surface of IBCs induced the observed effects at both high and low SAgAIns valency. Future studies aim to test the effects of SAgAIns on disease progression in the VH125.NOD mouse model of T1D.
Collapse
Affiliation(s)
- Martin A Leon
- Department of Chemistry , University of Kansas , 1251 Wescoe Hall Drive , Lawrence , Kansas 66045 , United States
| | - Scott M Wemlinger
- Department Immunology & Microbiology , University of Colorado Denver AMC , 12800 E. 19th Ave. , Aurora , Colorado 80045 , United States
| | - Nicholas R Larson
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| | - Justin K Ruffalo
- Department of Chemical and Petroleum Engineering , University of Kansas , 1530 West 15th Street , Lawrence , Kansas 66045 , United States
| | - Joshua O Sestak
- Orion BioScience , 986099 Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States
| | - John C Cambier
- Department Immunology & Microbiology , University of Colorado Denver AMC , 12800 E. 19th Ave. , Aurora , Colorado 80045 , United States
| | - Cory Berkland
- Department of Chemistry , University of Kansas , 1251 Wescoe Hall Drive , Lawrence , Kansas 66045 , United States.,Department of Pharmaceutical Chemistry , University of Kansas , 2095 Constant Avenue , Lawrence , Kansas 66047 , United States.,Department of Chemical and Petroleum Engineering , University of Kansas , 1530 West 15th Street , Lawrence , Kansas 66045 , United States
| |
Collapse
|
21
|
Arsiwala A, Castro A, Frey S, Stathos M, Kane RS. Designing Multivalent Ligands to Control Biological Interactions: From Vaccines and Cellular Effectors to Targeted Drug Delivery. Chem Asian J 2019; 14:244-255. [DOI: 10.1002/asia.201801677] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Ammar Arsiwala
- School of Chemical and Biomolecular Engineering; Georgia Institute of Technology; Atlanta Georgia- 30332 USA
| | - Ana Castro
- School of Chemical and Biomolecular Engineering; Georgia Institute of Technology; Atlanta Georgia- 30332 USA
| | - Steven Frey
- School of Chemical and Biomolecular Engineering; Georgia Institute of Technology; Atlanta Georgia- 30332 USA
| | - Mark Stathos
- School of Chemical and Biomolecular Engineering; Georgia Institute of Technology; Atlanta Georgia- 30332 USA
| | - Ravi S. Kane
- School of Chemical and Biomolecular Engineering; Georgia Institute of Technology; Atlanta Georgia- 30332 USA
| |
Collapse
|
22
|
Hartwell BL, Pickens CJ, Leon M, Northrup L, Christopher MA, Griffin JD, Martinez-Becerra F, Berkland C. Soluble antigen arrays disarm antigen-specific B cells to promote lasting immune tolerance in experimental autoimmune encephalomyelitis. J Autoimmun 2018; 93:76-88. [PMID: 30007842 PMCID: PMC6117839 DOI: 10.1016/j.jaut.2018.06.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/26/2022]
Abstract
Autoreactive lymphocytes that escape central immune tolerance may be silenced via an endogenous peripheral tolerance mechanism known as anergy. Antigen-specific therapies capable of inducing anergy may restore patients with autoimmune diseases to a healthy phenotype while avoiding deleterious side effects associated with global immunosuppression. Inducing anergy in B cells may be a particularly potent intervention, as B cells can contribute to autoimmune diseases through multiple mechanisms and offer the potential for direct antigen-specific targeting through the B cell receptor (BCR). Our previous results suggested autoreactive B cells may be silenced by multivalent 'soluble antigen arrays' (SAgAs), which are polymer conjugates displaying multiple copies of autoantigen with or without a secondary peptide that blocks intracellular cell-adhesion molecule-1 (ICAM-1). Here, key therapeutic molecular properties of SAgAs were identified and linked to the immunological mechanism through comprehensive cellular and in vivo analyses. We determined non-hydrolyzable 'cSAgAs' displaying multivalent 'click'-conjugated antigen more potently suppressed experimental autoimmune encephalomyelitis (EAE) compared to hydrolyzable SAgAs capable of releasing conjugated antigen. cSAgAs restored a healthy phenotype in disease-specific antigen presenting cells (APCs) by inducing an anergic response in B cells and a subset of B cells called autoimmune-associated B cells (ABCs) that act as potent APCs in autoimmune disease. Accompanied by a cytokine response skewed towards a Th2/regulatory phenotype, this generated an environment of autoantigenic tolerance. By identifying key therapeutic molecular properties and an immunological mechanism that drives SAgA efficacy, this work guides the design of antigen-specific immunotherapies capable of inducing anergy.
Collapse
MESH Headings
- Animals
- Autoantigens/genetics
- Autoantigens/immunology
- B-Lymphocyte Subsets/drug effects
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/pathology
- Click Chemistry
- Clonal Anergy/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Hydrolysis
- Immunoconjugates/chemistry
- Immunoconjugates/pharmacology
- Immunotherapy/methods
- Injections, Subcutaneous
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/immunology
- Mice
- Myelin Proteolipid Protein/administration & dosage
- Peptide Fragments/administration & dosage
- Peptide Fragments/chemical synthesis
- Peptide Fragments/immunology
- Peptide Fragments/pharmacology
- Protein Array Analysis
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Spleen/immunology
- Spleen/pathology
- Th2 Cells/immunology
- Th2 Cells/pathology
Collapse
Affiliation(s)
- Brittany L Hartwell
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA
| | - Chad J Pickens
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - Martin Leon
- Department of Chemistry, University of Kansas, 1251 Wescoe Hall Drive, Lawrence, KS 66045, USA
| | - Laura Northrup
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - Matthew A Christopher
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA
| | - J Daniel Griffin
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA
| | - Francisco Martinez-Becerra
- Immunology Core Laboratory of the Kansas Vaccine Institute, University of Kansas, 2030 Becker Drive, Lawrence, KS 66047, USA
| | - Cory Berkland
- Bioengineering Graduate Program, University of Kansas, 1520 West 15th Street, Lawrence, KS 66045, USA; Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA; Department of Chemical and Petroleum Engineering, University of Kansas, 1530 West 15th Street, Lawrence, KS 66045, USA.
| |
Collapse
|
23
|
Hartwell BL, Pickens CJ, Leon M, Berkland C. Multivalent Soluble Antigen Arrays Exhibit High Avidity Binding and Modulation of B Cell Receptor-Mediated Signaling to Drive Efficacy against Experimental Autoimmune Encephalomyelitis. Biomacromolecules 2017; 18:1893-1907. [PMID: 28474886 DOI: 10.1021/acs.biomac.7b00335] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A pressing need exists for antigen-specific immunotherapies (ASIT) that induce selective tolerance in autoimmune disease while avoiding deleterious global immunosuppression. Multivalent soluble antigen arrays (SAgAPLP:LABL), consisting of a hyaluronic acid (HA) linear polymer backbone cografted with multiple copies of autoantigen (PLP) and cell adhesion inhibitor (LABL) peptides, are designed to induce tolerance to a specific multiple sclerosis (MS) autoantigen. Previous studies established that hydrolyzable SAgAPLP:LABL, employing a degradable linker to codeliver PLP and LABL, was therapeutic in experimental autoimmune encephalomyelitis (EAE) in vivo and exhibited antigen-specific binding with B cells, targeted the B cell receptor (BCR), and dampened BCR-mediated signaling in vitro. Our results pointed to sustained BCR engagement as the SAgAPLP:LABL therapeutic mechanism, so we developed a new version of the SAgA molecule using nonhydrolyzable conjugation chemistry, hypothesizing it would enhance and maintain the molecule's action at the cell surface to improve efficacy. "Click SAgA" (cSAgAPLP:LABL) uses hydrolytically stable covalent conjugation chemistry (Copper-catalyzed Azide-Alkyne Cycloaddition (CuAAC)) rather than a hydrolyzable oxime bond to attach PLP and LABL to HA. We explored cSAgAPLP:LABL B cell engagement and modulation of BCR-mediated signaling in vitro through flow cytometry binding and calcium flux signaling assays. Indeed, cSAgAPLP:LABL exhibited higher avidity B cell binding and greater dampening of BCR-mediated signaling than hydrolyzable SAgAPLP:LABL. Furthermore, cSAgAPLP:LABL exhibited significantly enhanced in vivo efficacy compared to hydrolyzable SAgAPLP:LABL, achieving equivalent efficacy at one-quarter of the dose. These results indicate that nonhydrolyzable conjugation increased the avidity of cSAgAPLP:LABL to drive in vivo efficacy through modulated BCR-mediated signaling.
Collapse
Affiliation(s)
- Brittany L Hartwell
- Bioengineering Graduate Program, University of Kansas 1520 West 15th Street, Lawrence, Kansas 66045, United States
| | - Chad J Pickens
- Department of Pharmaceutical Chemistry, University of Kansas 2095 Constant Avenue, Lawrence, Kansas 66047, United States
| | - Martin Leon
- Department of Chemistry, University of Kansas 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| | - Cory Berkland
- Bioengineering Graduate Program, University of Kansas 1520 West 15th Street, Lawrence, Kansas 66045, United States.,Department of Pharmaceutical Chemistry, University of Kansas 2095 Constant Avenue, Lawrence, Kansas 66047, United States.,Department of Chemical and Petroleum Engineering, University of Kansas 1530 West 15th Street, Lawrence, Kansas 66045, United States
| |
Collapse
|