Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1258369, 2024.
Article in English | MEDLINE | ID: mdl-38933266

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.


Subject(s)
Diabetes Mellitus, Type 1 , Immune Tolerance , Mice, Inbred NOD , Peptides , Animals , Mice , Diabetes Mellitus, Type 1/immunology , Peptides/immunology , Peptides/administration & dosage , Female , Autoantigens/immunology , T-Lymphocytes, Regulatory/immunology , Immunotherapy/methods , Anaphylaxis/prevention & control , Anaphylaxis/immunology , Desensitization, Immunologic/methods , Desensitization, Immunologic/adverse effects
2.
bioRxiv ; 2023 May 07.
Article in English | MEDLINE | ID: mdl-37205572

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.

3.
Diabetes ; 70(6): 1334-1346, 2021 06.
Article in English | MEDLINE | ID: mdl-33468513

ABSTRACT

Antigen-specific immunotherapy (ASIT) offers a targeted treatment of autoimmune diseases that selectively inhibits autoreactive lymphocytes, but there remains an unmet need for approaches that address the limited clinical efficacy of ASIT. Soluble antigen arrays (SAgAs) deliver antigenic peptides or proteins in multivalent form, attached to a hyaluronic acid backbone using either hydrolysable linkers (hSAgAs) or stable click chemistry linkers (cSAgAs). They were evaluated for the ability to block spontaneous development of disease in a nonobese diabetic mouse model of type 1 diabetes (T1D). Two peptides, a hybrid insulin peptide and a mimotope, efficiently prevented the onset of T1D when delivered in combination as SAgAs, but not individually. Relative to free peptides administered at equimolar dose, SAgAs (particularly cSAgAs) enabled a more effective engagement of antigen-specific T cells with greater persistence and induction of tolerance markers, such as CD73, interleukin-10, programmed death-1, and KLRG-1. Anaphylaxis caused by free peptides was attenuated using hSAgA and obviated using cSAgA platforms. Despite similarities, the two peptides elicited largely nonoverlapping and possibly complementary responses among endogenous T cells in treated mice. Thus, SAgAs offer a novel and promising ASIT platform superior to free peptides in inducing tolerance while mitigating risks of anaphylaxis for the treatment of T1D.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Peptides/pharmacokinetics , Protein Array Analysis , Animals , Autoantigens/immunology , Click Chemistry , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/immunology , Drug Delivery Systems , Drug Liberation , Female , Immunotherapy/instrumentation , Immunotherapy/methods , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Peptides/administration & dosage , Remission Induction/methods , Solubility , Treatment Outcome
4.
Adv Drug Deliv Rev ; 165-166: 105-116, 2020.
Article in English | MEDLINE | ID: mdl-32325104

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.


Subject(s)
Autoantigens/immunology , Drug Delivery Systems/methods , Immunotherapy/methods , Allergens/chemistry , Allergens/pharmacology , Autoimmune Diseases/physiopathology , Biological Transport/physiology , Desensitization, Immunologic/methods , Drug Administration Routes , Humans
5.
ACS Chem Biol ; 14(7): 1436-1448, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31260253

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.


Subject(s)
Diabetes Mellitus, Type 1/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation , Cells, Cultured , Diabetes Mellitus, Type 1/therapy , Epitopes/immunology , Female , Immunotherapy, Adoptive , Mice, Inbred NOD , Mice, Transgenic , Protein Array Analysis , Spleen/immunology
6.
Biomacromolecules ; 20(5): 2115-2122, 2019 05 13.
Article in English | MEDLINE | ID: mdl-30995843

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.


Subject(s)
Autoantigens/chemistry , B-Lymphocytes/immunology , Immunomodulation , Spleen/cytology , Animals , Autoantigens/immunology , B7-2 Antigen/immunology , Cell Line, Tumor , Cells, Cultured , Click Chemistry/methods , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Immunoassay/methods , Mice , Protein Array Analysis/methods , Spleen/immunology
7.
Mol Pharm ; 16(4): 1563-1572, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30681867

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.


Subject(s)
Autoantigens/immunology , B-Lymphocytes/immunology , Insulin/immunology , Multiple Sclerosis/immunology , Peptide Fragments/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Autoantigens/metabolism , B-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Female , Humans , Hyaluronic Acid/chemistry , Immune Tolerance , Insulin/metabolism , Mice , Mice, Inbred NOD , Multiple Sclerosis/metabolism , Peptide Fragments/metabolism , Protein Array Analysis , Receptors, Antigen, B-Cell/metabolism
8.
Mol Pharm ; 13(2): 330-43, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26636828

ABSTRACT

Many current therapies for autoimmune diseases such as multiple sclerosis (MS) result in global immunosuppression, rendering insufficient efficacy with increased risk of adverse side effects. Multivalent soluble antigen arrays, nanomaterials presenting both autoantigen and secondary inhibitory signals on a flexible polymer backbone, are hypothesized to shift the immune response toward selective autoantigenic tolerance to repress autoimmune disease. Two-signal co-delivery of both autoantigen and secondary signal were deemed necessary for therapeutic efficacy against experimental autoimmune encephalomyelitis, a murine model of MS. Dynamic light scattering and in silico molecular dynamics simulations complemented these studies to illuminate the role of two-signal co-delivery in determining therapeutic potential. Physicochemical characteristics such as particle size and molecular affinity for intermolecular interactions and chain entanglement likely facilitated cotransport of two signals to produce efficacy. These findings elucidate potential mechanisms whereby soluble antigen arrays enact their therapeutic effect and help to guide the development of future multivalent antigen-specific immunotherapies.


Subject(s)
Autoantigens/immunology , Drug Delivery Systems , Encephalomyelitis, Autoimmune, Experimental/therapy , Immune Tolerance/immunology , Nanostructures/chemistry , Polymers/chemistry , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunotherapy , Mice , Molecular Dynamics Simulation , Protein Array Analysis
9.
J Pharm Sci ; 104(2): 346-61, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25447598

ABSTRACT

Continued development of multivalent nanomaterials has provided opportunities for the advancement of antigen-specific immunotherapies. New insights emerge when considering the backdrop of vaccine design, which has long employed multivalent presentation of antigen to more strongly engage and enhance an immunogenic response. Additionally, vaccines traditionally codeliver antigen with adjuvant to amplify a robust antigen-specific response. Multivalent nanomaterials have since evolved for applications where immune tolerance is desired, such as autoimmune diseases or allergies. In particular, soluble, linear polymers may be tailored to direct antigen-specific immunogenicity or tolerance by modulating polymer length, ligand valency (number), and ligand density, in addition to incorporating secondary signals. Codelivery of a secondary signal may direct, amplify, or suppress the response to a given antigen. Although the ability of multivalent nanomaterials to enact an immune response through molecular mechanisms has been established, a transport mechanism for biodistribution must also be considered. Both mechanisms are influenced by ligand display and other physical properties of the nanomaterial. This review highlights multivalent ligand display on linear polymers, the complex interplay of physical parameters in multivalent design, and the ability to direct the immune response by molecular and transport mechanisms.


Subject(s)
Antigens/immunology , Immunotherapy , Nanostructures/chemistry , Nanostructures/therapeutic use , Vaccines/chemistry , Vaccines/immunology , Humans , Ligands , Polymers/chemistry
10.
AAPS J ; 16(6): 1204-13, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25297853

ABSTRACT

Autoimmune diseases such as multiple sclerosis (MS) are characterized by the breakdown of immune tolerance to autoantigens. Targeting surface receptors on immune cells offers a unique strategy for reprogramming immune responses in autoimmune diseases. The B7 signaling pathway was targeted using adaptations of soluble antigen array (SAgA) technology achieved by covalently linking B7-binding peptides and disease causing autoantigen (proteolipid peptide (PLP)) to hyaluronic acid (HA). We hypothesized that co-delivery of a B7-binding peptide and autoantigen would suppress experimental autoimmune encephalomyelitis (EAE), a murine model of MS. Three independent B7-targeted SAgAs were created containing peptides to either inhibit or potentially stimulate the B7 signaling pathway. Surprisingly, all SAgAs were found to suppress EAE disease symptoms. Altered cytokine expression was observed in primary splenocytes isolated from SAgA-treated mice, indicating that SAgAs with different B7-binding peptides may suppress EAE through different immunological mechanisms. This antigen-specific immunotherapy using SAgAs can successfully suppress EAE through co-delivery of autoantigen and peptides targeting with the B7 signaling pathway.


Subject(s)
Autoantigens/immunology , B7 Antigens/immunology , Drug Carriers/chemistry , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immune Tolerance/drug effects , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Peptides/therapeutic use , Animals , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Hyaluronic Acid/chemistry , Immune Tolerance/immunology , Mice, Inbred Strains , Peptides/administration & dosage , Peptides/chemistry , Solubility , Spleen/cytology , Spleen/drug effects , Spleen/immunology
11.
AAPS J ; 16(6): 1185-93, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25193268

ABSTRACT

Presentation of antigen with immune stimulating "signal" has been a cornerstone of vaccine design for decades. Here, the antigen plus immune "signal" of vaccines is modified to produce antigen-specific immunotherapies (antigen-SITs) that can potentially reprogram the immune response toward tolerance of an autoantigen. The codelivery of antigen with a cell adhesion inhibitor using Soluble Antigen Arrays (SAgAs) was previously shown to slow or halt experimental autoimmune encephalomyelitis (EAE), a murine form of multiple sclerosis (MS). SAgAs are comprised of a hyaluronic acid backbone with cografted intercellular cell adhesion molecule-1 ligand derived from αL-integrin (CD11a237-246, "LABL") and an encephalitogenic epitope peptide of proteolipid protein (PLP139-151, "PLP"). Here, the physical characteristics of the carrier were investigated to evaluate how structure, size, and solubility drive the immune response when treating EAE. A bifunctional peptide (small, soluble), SAgAs (large, soluble), and PLGA nanoparticles (large, insoluble) all displaying PLP and LABL in equimolar ratios were compared. Maximum EAE suppression was achieved with coincident display of both peptides on a soluble construct.


Subject(s)
Autoantigens/immunology , Drug Carriers/chemistry , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immune Tolerance/drug effects , Peptides/therapeutic use , Animals , CD11a Antigen/immunology , Drug Compounding , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Hyaluronic Acid/chemistry , Lactic Acid/chemistry , Mice, Inbred Strains , Myelin Proteolipid Protein/immunology , Nanoparticles/chemistry , Particle Size , Peptide Fragments/immunology , Peptides/administration & dosage , Peptides/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Solubility
12.
Mol Ther Methods Clin Dev ; 1: 14008, 2014.
Article in English | MEDLINE | ID: mdl-26015953

ABSTRACT

Autoimmune diseases such as multiple sclerosis (MS) are typified by the misrecognition of self-antigen and the clonal expansion of autoreactive T cells. Antigen-specific immunotherapies (antigen-SITs) have long been explored as a means to desensitize patients to offending self-antigen(s) with the potential to retolerize the immune response. Soluble antigen arrays (SAgAs) are composed of hyaluronic acid (HA) cografted with disease-specific autoantigen (proteolipid protein peptide) and an ICAM-1 inhibitor peptide (LABL). SAgAs were designed as an antigen-SIT that codeliver peptides to suppress experimental autoimmune encephalomyelitis (EAE), a murine model of MS. Codelivery of antigen and cell adhesion inhibitor (LABL) conjugated to HA was essential for SAgA treatment of EAE. Individual SAgA components or mixtures thereof reduced proinflammatory cytokines in cultured splenocytes from EAE mice; however, these treatments showed minimal to no in vivo therapeutic effect in EAE mice. Thus, carriers that codeliver antigen and a secondary "context" signal (e.g., LABL) in vivo may be an important design criteria to consider when designing antigen-SIT for autoimmune therapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...