scholarly journals 779. Oral Delivery of Non-Viral DNA Nanoparticles for Hemophilia A

2005 ◽  
Vol 11 ◽  
pp. S302
Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 14-14 ◽  
Author(s):  
Roland W Herzog ◽  
Dheeraj Verma ◽  
Xiaomei Wang ◽  
Alexandra Sherman ◽  
Shina Lin ◽  
...  

Abstract Abstract 14 Approximately 25% of hemophilia A patients develop inhibitors against factor VIII during replacement therapy by infusion of factor VIII concentrates, rendering this treatment ineffective. Elimination of this antibody response is currently achieved by highly expensive immune tolerance induction (ITI) protocols involving prolonged administration of FVIII. No prophylactic immune tolerance protocols are available. To overcome these limitations, this study seeks to develop a cost-effective approach for tolerance induction by oral delivery of human factor VIII (hF.VIII) immunogenic domains expressed in chloroplasts and bioencapsulated in plant cells. Previously, we have shown that this approach effectively suppresses inhibitor formation and anaphylaxis against factor IX in hemophilia B mice (PNAS 107:7101, 2010). Bioencapsulation protects protein antigens from gastric enzymes and acidic environment of the stomach, resulting in antigen release to the immune system via digestion of plant cell walls by microbes that colonizing the gut. The transplastomic tobacco plants created expressed the heavy chain (HC, A1-A2), A3 and C2 domains fused to the transmucosal carrier cholera toxin B subunit (CTB) to facilitate GM1 receptor mediated delivery. Besides a GPGP hinge, a furin cleavage site was introduced to link CTB with the different domains of hF.VIII coding sequence for proper folding and release of hF.VIII domains into the circulatory or immune system. PCR and/or Southern blot analysis was carried out to confirm site-specific transgene integration. Western blot analysis showed expected size fusion protein band in all four transplastomic lines expressing CTB-HC, CTB-A2, CTB-A3 and CTB-C2 fusion protein. The GM1-ganglioside receptor binding ELISA assay with chloroplast synthesized CTB-C2 and CTB-A2 fusion protein showed equivalent absorbance when compared to the purified CTB, confirming the correct folding and disulfide-bond formation of CTB pentamers within transformed chloroplasts. Transplastomic leaves expressed CTB-HC, CTB-A2, CTB-A3 and CTB-C2 in the range of 0.4–1.4%, 0.1–0.2%, 0.3–0.7% and 3.0–9.1% in the total leaf protein. Leaf materials were ground in liquid nitrogen and orally delivered to male hemophilia A mice (C57BL6/129 F8e16 −/−) for tolerance induction. In a first set of experiments, 125 mg plant material was used per oral dose, representing a mix of an approximately equal amount of HC-CTB and C2-CTB fusion proteins. Gavages were performed twice per week for 8 weeks. Control mice were fed with wild-type plant material. During the last 4 weeks, all mice (n=6 per group) were additionally treated with recombinant B domain deleted human F.VIII (intravenous injection of 1 IU once per week). By the end of the experiment, control mice had formed IgG2a (up to 0.9 μg/ml) and IgG2b (up to 1.7 μg/ml) titers against hFVIII, which were undetectable in hF.VIII-fed mice. Moreover, the control mice formed very high-titer IgG1 against hF.VIII (ranging from 7–24 μg/ml), resulting in an inhibitor titer of up to 400 BU (with an average of 211±126 BU). In contrast, hFVIII-fed mice only developed 1.9±0.6 μg/ml IgG1 and 30±12 BU, representing a highly significant (P=0.006 and P=0.001, respectively) 7–10 fold reduction in antibody formation upon factor replacement therapy. These data demonstrate that hF.VIII antigen can be produced by transplastomic technology and provide first proof-of-principle that oral delivery of bioencapsulated hF.VIII antigen is effective in controlling inhibitor development. Current work focuses on further optimization, and generation of an edible crop plant (lettuce) expressing hFVIII domains in the chloroplast for future translational studies is well on its way. Disclosures: Daniell: Bayer: Research Funding.


2004 ◽  
Vol 10 (1) ◽  
pp. 112 ◽  
Author(s):  
Jing Chen ◽  
Wu-Li Yang ◽  
Ge Li ◽  
Ji Qian ◽  
Jing-Lun Xue ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2563-2563
Author(s):  
Davide Matino ◽  
Marco Gargaro ◽  
Giulia Scalisi ◽  
Antonella Turco ◽  
Francisco Javier Quintana ◽  
...  

Abstract Background: The development of inhibitors is the most serious complication a patient with hemophilia can experience on treatment with clotting factor concentrates. In a cohort of 100 severe hemophilia A patients, we have recently found that the inhibitor-positive status was associated with dysfunctional indoleamine 2,3-dioxygenase 1 (IDO1)1. The mechanisms whereby IDO1 promotes regulatory effects include production of tryptophan catabolites, collectively known as kynurenines. Some of those tryptophan derivatives are endogenous in nature and act as activating ligands for the Aryl hydrocarbon receptor (AhR), a transcription factor that belongs in the family of basic helix-loop-helix transcription factors2. Specifically, in dendritic cells (DCs), tryptophan-catabolite interactions with AhR promote the transcriptional induction of anti-inflammatory cytokines and the emergence of T regulatory (Treg) cells. Here, we report on the potential of tryptophan-related AhR ligands for inhibiting the formation of anti-FVIII antibodies in hemophilic (F8 KO) mice, a finding that could be important in developing novel strategies to prevent or eradicate FVIII inhibitors. Methods: Four different endogenous AhR ligands have been selected and used in these experiments. All molecules were administered in vivo to F8-deficient mice at incremental doses and by different routes (orally or intravenously), either concomitantly or after rhFVIII immunization. In specific experiments, AhR knockout mice and co-administration of AhR antagonist were used to analyze the impact of AhR deficiency. To co-deliver AhR ligands and FVIII antigens to APCs in vivo and induce antigen-specific Tregs, we administered molecules via a nanostructured delivery system3. Specifically, we engineered four types of nanoparticles (NPs), 60 nm in diameter, that were stabilized by a thiol-polyethyleneglycol (PEG) layer. Overall, we used 1) unloaded NPs, 2) NPs loaded with AhR ligands, 3) NPs loaded with rhFVIII, and 4) NPs loaded with AhR ligands and rhFVIII. Results: Administration of one of the four AhR ligands prevented the generation of anti-FVIII antibodies in nearly 80% of F8 KO mice. Similar effects were obtained with both the NPs-based approach and the oral delivery of ligands. Of note, the protective effect was negated by co-administration of the AhR antagonist CH-223191. Conclusions: Our results suggest that AhR is a possible molecular partner whereby tryptophan catabolites will control the immune response to FVIII. These findings might lead to novel interventions for preventing or eradicating inhibitors in hemophilia A patients. Bibliography: 1.Matino D., et al. IDO1 suppresses inhibitor development in hemophilia A treated with factor VIII. J Clin Invest. 2015 Oct 1;125(10):3766-81. 2. Yeste A, Nadeau M, Burns EJ, Weiner HL, Quintana FJ. Nanoparticle-mediated codelivery of myelin antigen and a tolerogenic small molecule suppresses experimental autoimmune encephalomyelitis. Proc. Natl Acad. Sci. USA 109(28), 11270-11275 (2012). 3. Bessede A, et al. Aryl hydrocarbon receptor control of a disease tolerance defence pathway.Nature. 2014;511(7508):184-190. Disclosures No relevant conflicts of interest to declare.


2008 ◽  
Vol 132 (3) ◽  
pp. 252-259 ◽  
Author(s):  
Katherine Bowman ◽  
Rita Sarkar ◽  
Sanj Raut ◽  
Kam W. Leong

Blood ◽  
2014 ◽  
Vol 124 (10) ◽  
pp. 1659-1668 ◽  
Author(s):  
Alexandra Sherman ◽  
Jin Su ◽  
Shina Lin ◽  
Xiaomei Wang ◽  
Roland W. Herzog ◽  
...  

Key Points Factor VIII antigens can be expressed in chloroplasts and bioencapsulated in plant cells. Oral delivery of plant cells expressing FVIII domains suppresses and reverses inhibitor formation in mice with hemophilia A by induction of CD4+ regulatory T cells.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 8-9
Author(s):  
Thais Bertolini ◽  
Moanaro Biswas ◽  
Radoslaw Kaczmarek ◽  
Henry Daniell ◽  
Roland W Herzog

A serious complication in the treatment X-linked bleeding disorder hemophilia A is the formation of inhibitory antibodies against factor VIII (FVIII), which compromise traditional replacement therapy. We previously developed an Oral immunotherapy (OTI) based on repeated uptake of a mixture of lettuce plant cells transgenic for heavy chain (HC) or C2 domain of human FVIII fused to cholera toxin B (CTB) subunit [Blood 124:1659; Plant Biotechnol J. 16:1148]. Fusion proteins were transgenically expressed in the chloroplasts. Repeated oral uptake of a mixture of freeze-dried powder of lettuce cells accomplished antigen delivery to the immune system of the small intestine by targeting of the GM1 receptor that is highly expressed on the surface of the gut epithelium, resulting in induction of regulatory T cells (Treg) that suppress inhibitor formation upon subsequent intravenous (iv) FVIII replacement therapy. An alternative to oral antigen delivery is the oral delivery of immune modulatory antibodies. Here, we compared the plant cell-based method with oral delivery of anti-CD3, which has been successful in murine models of autoimmune disease and is currently evaluated in clinical trials. Unlike in iv administration, oral anti-CD3 does not systemically deplete T cells. Hemophilia A BALB/c mice (F8 e16 gene deletion) received oral gavage of a mixture of CTB-FVIII-HC/-C2 (1.5 µg/antigen) expressing lettuce leaf cells 2x/week for 9 weeks. Starting at 4 weeks into the experiment, 1 IU/mouse of BDD-FVIII (Xyntha) was given iv, once per week for 5 weeks. Alternatively, following a published protocol that was successful in other models, 5 µg of hamster anti-murine CD3 was given by oral gavage daily for 5 straight days, followed by 5 weekly iv injections of BDD-FVIII. Control animals (no OTI) developed inhibitors with an average titer of 18 ± 3 BU/ml (n=16). Of these, 88% were high-titer (i.e >5 BU/ml, up to 43 BU/ml). Inhibitor formation was significantly reduced in plant cell-treated mice (10 ± 2.5 BU/ml, n=17), with 47% showing no or low-titer inhibitors (<5 BU/ml). Interestingly, inhibitor formation in anti-CD3 treated mice showed a unique pattern, with two distinct outcomes. One subset of animals developed inhibitors of 0-15 BU/ml (on average 2.5-fold lower than control group), while the second subset showed increased titers of 41-89 BU/ml. There were no animals with intermediate titers. In general, FVIII-specific IgG titers followed the same patterns as inhibitor responses. When the 2 approaches (plant cells and anti-CD3) were combined, 50% of mice developed <5 BU/ml (as compared to 36% for anti-CD3 only). However, this did not represent an improvement over giving lettuce cells only. Finally, we tested anti-CD3 Fab fragment in place of FL anti-CD3. While a daily dose of 5 µg anti-CD3 Fab/mouse was unsuccessful in suppressing inhibitor formation, a dose of 0.5 µg marginally decrease titers. In summary, oral anti-CD3 delivery was not as effective in reducing inhibitor formation as plant cells expressing CTB-FVIII antigen fusions. While FL anti-CD3 showed promise in a subset of animals, it also posed a risk of an increased response in others, which complicates development of this approach. Distinct from all other treatment groups, lettuce-fed mice showed elevated frequencies in Foxp3+ Treg, a reduction in Teff cells, and the highest increase in LAP+ Treg. Further studies on induced CD4+LAP+FoxP3- Treg cells from lettuce-fed mice showed high expression of ICOS, CD69 and Ki-67, but not CTLA-4. Among the different subsets of Treg, LAP+ T cell contained the highest proportion of IL-10 expressing cells, and oral antigen delivery induced IL-10+LAP+ Treg, which are likely key to the suppression of the immune response against FVIII. Disclosures Daniell: Takeda Pharmaceuticals: Patents & Royalties. Herzog:Takeda Pharmaceuticals: Patents & Royalties.


Sign in / Sign up

Export Citation Format

Share Document