The undesired destruction of healthy cells by the immune system results in the loss of tissue function and complicates strategies to restore tissue function. The current standard therapy for autoimmune disease involves generalized immunosuppression, which is in most cases is not clinically efficacious and leads to numerous undesired side effects. Dr. Stephen Miller, (co-PI) pioneered an approach in which splenocytes were cross-linked with specific auto antigens, and their delivery to the spleen induced tolerance specifically to the auto antigen. This approach was recently adapted for a clinical trial in multiple sclerosis (MS) patients, and was the first-in-man study to report the induction antigen specific tolerance. However, the use of cellular carriers for tolerance induction in the clinical arena is challenging due to the considerable ex-vivo laboratory manipulation that is required, which is expensive, increases the number of donor cells needed and introduces further opportunity for technical error. Our long-term goal is to develop a particle-based platform that can be an off-the-shelf product for induction of tolerance to specific antigens to inhibit the specific undesired immune response while not altering the remaining elements of the immune response. We have demonstrated that antigen-loaded particles delivered intravenously can induce tolerance for the prevention and treatment of experimental autoimmune encephalomyelitis (EAE), the mouse model of MS. With the goal of moving this technology toward the clinic, we propose to extend these studies to address fundamental questions about the particle design and their mechanisms of action, and also critical questions regarding the ability to target the variety of antigens and cell populations underlying disease.
Specific Aim 1 will investigate the particle design parameters and identify the cellular mechanisms by which particles injected intravenously are able to modulate inflammation and induce antigen specific tolerance. Our results suggest the liver as a critical site involved in tolerance induction from the particles, which distinguishes it from the previous work with antigen-coupled splenocytes. We propose to investigate the particle composition and size to distinguish i) the impact of the carrier on immune cell polarization, ii) te efficacy of antigen presentation, and iii) in vivo trafficking of the particles.
Specific Aim 2 wil determine the cellular and molecular mechanisms by which Ag-PLG tolerance is induced and maintained in nave, activated, and memory T cells. We propose to test the ability to induce tolerance with particles encapsulating multiple peptides/proteins and to examine the separate and combined contributions of anergy and Tregs to the induction and maintenance of tolerance. Successful completion of these studies would identify particles that are novel, safe, efficient and clinically relevant tools to inhibit antigen-specific T-cells for therapy of autoimmune diseases. This innovative approach has far reaching implications for decreasing specific immune responses in applications such as autoimmune disease, rejection of transplanted cells, and allergies to food antigens or airborne particulates.

Public Health Relevance

In autoimmune diseases, such as multiple sclerosis, the immune system attacks healthy cells resulting in the loss of tissue function, and also complicates strategies to restore cells that could provide that function. We are designing biodegradable, biocompatible particles loaded with proteins that can induce tolerance to those proteins and thereby specifically prevent the attack by cells of the immune system. This innovative approach has far reaching implications for applications in which decreasing specific immune responses could be beneficial, such as the autoimmune diseases, rejection of transplanted cells in regenerative medicine, and allergies to food antigens or airborne particulates.

Agency
National Institute of Health (NIH)
Institute
National Institute of Biomedical Imaging and Bioengineering (NIBIB)
Type
Research Project (R01)
Project #
5R01EB013198-08
Application #
9235147
Study Section
Gene and Drug Delivery Systems Study Section (GDD)
Program Officer
Rampulla, David
Project Start
2011-04-01
Project End
2019-02-28
Budget Start
2017-03-01
Budget End
2018-02-28
Support Year
8
Fiscal Year
2017
Total Cost
$473,930
Indirect Cost
$77,805
Name
University of Michigan Ann Arbor
Department
Biomedical Engineering
Type
Schools of Engineering
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Prasad, Suchitra; Neef, Tobias; Xu, Dan et al. (2018) Tolerogenic Ag-PLG nanoparticles induce tregs to suppress activated diabetogenic CD4 and CD8 T cells. J Autoimmun 89:112-124
Skoumal, Michael; Woodward, Kyle B; Zhao, Hong et al. (2018) Localized immune tolerance from FasL-functionalized PLG scaffolds. Biomaterials 192:271-281
Neef, Tobias; Miller, Stephen D (2017) Tolerogenic Nanoparticles to Treat Islet Autoimmunity. Curr Diab Rep 17:84
Pearson, Ryan M; Casey, Liam M; Hughes, Kevin R et al. (2017) Controlled Delivery of Single or Multiple Antigens in Tolerogenic Nanoparticles Using Peptide-Polymer Bioconjugates. Mol Ther 25:1655-1664
Kuo, Robert; Saito, Eiji; Miller, Stephen D et al. (2017) Peptide-Conjugated Nanoparticles Reduce Positive Co-stimulatory Expression and T Cell Activity to Induce Tolerance. Mol Ther 25:1676-1685
Pearson, Ryan M; Casey, Liam M; Hughes, Kevin R et al. (2017) In vivo reprogramming of immune cells: Technologies for induction of antigen-specific tolerance. Adv Drug Deliv Rev 114:240-255
Jeong, Su Ji; Cooper, John G; Ifergan, Igal et al. (2017) Intravenous immune-modifying nanoparticles as a therapy for spinal cord injury in mice. Neurobiol Dis 108:73-82
McCarthy, Derrick P; Yap, Jonathan Woon-Teck; Harp, Christopher T et al. (2017) An antigen-encapsulating nanoparticle platform for TH1/17 immune tolerance therapy. Nanomedicine 13:191-200
Smarr, Charles B; Yap, Woon Teck; Neef, Tobias P et al. (2016) Biodegradable antigen-associated PLG nanoparticles tolerize Th2-mediated allergic airway inflammation pre- and postsensitization. Proc Natl Acad Sci U S A 113:5059-64
Ifergan, Igal; Chen, Siqi; Zhang, Bin et al. (2016) Cutting Edge: MicroRNA-223 Regulates Myeloid Dendritic Cell-Driven Th17 Responses in Experimental Autoimmune Encephalomyelitis. J Immunol 196:1455-1459

Showing the most recent 10 out of 28 publications