scholarly journals Antigen-specific immunotherapy with apitopes suppresses generation of FVIII inhibitor antibodies in HLA-transgenic mice

Author(s):  
Katrien Pletinckx ◽  
Kirsty S Nicolson ◽  
Heather B Streeter ◽  
William J Sanderson ◽  
Evelien Schurgers ◽  
...  

Haemophilia A (HA) is a blood clotting disorder caused by various genetic deficiencies in the factor VIII (FVIII) encoding F8 gene. Patients receiving FVIII replacement therapy are at risk of developing neutralizing antibodies (FVIII inhibitors) rendering the FVIII replacement therapy ineffective. Immunological tolerance towards FVIII can be achieved through immune tolerance induction (ITI) protocols in some patients but this is a lengthy and costly desensitization programme. Long-term eradication of inhibitors in HA patients could be achieved by antigen-specific immunotherapy targeting CD4+ T cells since formation of FVIII inhibitors is T cell dependent. Here, we report a peptide-based, antigen-specific immunotherapy designed to specifically re-establish immune tolerance to FVIII through the development of antigen-processing-independent epitopes (apitopes). We identified two FVIII immunodominant peptides in immunised human leukocyte antigen (HLA) DRA*0101/DRB1*1501 transgenic (HLA-DR2tg) mice that were optimised for tolerogenicity. These modified peptide analogues were initially screened for recognition using FVIII-specific T cell hybridoma clones from FVIII-immunised HLA-DR2tg mice. The FVIII apitopes were promiscuous and bound common human HLA-DRB1*haplotypes. The combination of these two FVIII apitopes (ATX-F8-117), administered according to a dose escalation protocol, promoted T cell tolerance towards FVIII in HLA-DR2tg mice. Furthermore, treatment with ATX-F8-117 significantly reduced FVIII inhibitor formation. ATX-F8-117 regulates both anti-FVIII T cell and B cell responses, specifically the generation of FVIII inhibitors, revealing peptide-based antigen-specific immunotherapy as a promising approach to both suppress and treat inhibitor formation in susceptible HA patients.

2021 ◽  
Vol 12 ◽  
Author(s):  
Evelien Schurgers ◽  
David C. Wraith

The immune response to exogenous proteins can overcome the therapeutic benefits of immunotherapies and hamper the treatment of protein replacement therapies. One clear example of this is haemophilia A resulting from deleterious mutations in the FVIII gene. Replacement with serum derived or recombinant FVIII protein can cause anti-drug antibodies in 20-50% of individuals treated. The resulting inhibitor antibodies override the benefit of treatment and, at best, make life unpredictable for those treated. The only way to overcome the inhibitor issue is to reinstate immunological tolerance to the administered protein. Here we compare the various approaches that have been tested and focus on the use of antigen-processing independent T cell epitopes (apitopes) for tolerance induction. Apitopes are readily designed from any protein whether this is derived from a clotting factor, enzyme replacement therapy, gene therapy or therapeutic antibody.


2021 ◽  
Vol 12 ◽  
Author(s):  
Gabriel Goncalves ◽  
Kerry A. Mullan ◽  
Divya Duscharla ◽  
Rochelle Ayala ◽  
Nathan P. Croft ◽  
...  

Peptide vaccination remains a viable approach to induce T-cell mediated killing of tumors. To identify potential T-cell targets for Triple-Negative Breast Cancer (TNBC) vaccination, we examined the effect of the pro-inflammatory cytokine interferon-γ (IFNγ) on the transcriptome, proteome, and immunopeptidome of the TNBC cell line MDA-MB-231. Using high resolution mass spectrometry, we identified a total of 84,131 peptides from 9,647 source proteins presented by human leukocyte antigen (HLA)-I and HLA-II alleles. Treatment with IFNγ resulted in a remarkable remolding of the immunopeptidome, with only a 34% overlap between untreated and treated cells across the HLA-I immunopeptidome, and expression of HLA-II only detected on treated cells. IFNγ increased the overall number, diversity, and abundance of peptides contained within the immunopeptidome, as well increasing the coverage of individual source antigens. The suite of peptides displayed under conditions of IFNγ treatment included many known tumor associated antigens, with the HLA-II repertoire sampling 17 breast cancer associated antigens absent from those sampled by HLA-I molecules. Quantitative analysis of the transcriptome (10,248 transcripts) and proteome (6,783 proteins) of these cells revealed 229 common proteins and transcripts that were differentially expressed. Most of these represented downstream targets of IFNγ signaling including components of the antigen processing machinery such as tapasin and HLA molecules. However, these changes in protein expression did not explain the dramatic modulation of the immunopeptidome following IFNγ treatment. These results demonstrate the high degree of plasticity in the immunopeptidome of TNBC cells following cytokine stimulation and provide evidence that under pro-inflammatory conditions a greater variety of potential HLA-I and HLA-II vaccine targets are unveiled to the immune system. This has important implications for the development of personalized cancer vaccination strategies.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2188-2188
Author(s):  
Christine Lenk ◽  
Markus Pasztorek ◽  
Markus Weiller ◽  
Rafi Uddin Ahmad ◽  
Hans Peter Schwarz ◽  
...  

Abstract Abstract 2188 25%-30% of patients with hemophilia A develop neutralizing antibodies following replacement therapy with factor VIII (FVIII). These patients can be treated with factor VIIa (FVIIa) which triggers the extrinsic pathway of coagulation and thereby bypasses the requirement for FVIII. We developed a new mouse model that is transgenic for human FVII and expresses specific immune tolerance to native human FVIIa. We aim to investigate the immunological impact of modified FVIIa product candidates and to characterize their immunogenicity by analyzing emerging FVIIa-specific T cell responses. The new mouse model offers a unique opportunity to study central and peripheral immune regulatory mechanisms and the generation of immune responses by pro-inflammatory antigen-specific effector T cells (Teff). We hypothesized that FVIIa-specific Teff having escaped clonal deletion are present in the periphery and may be actively suppressed by FVIIa-specific regulatory T cells (Tregs). To study this hypothesis, we immunized mice with recombinant FVIIa (rFVIIa) with or without LPS, a well-described “danger signal” being able to break immune tolerance by stimulating the innate immune system. Intravenous or subcutaneous administration of rFVIIa alone did not elicit antibody responses and thus immune tolerance to rFVIIa was not broken. However, co-administration of rFVIIa and LPS resulted in a specific antibody response that was not isotypically restricted. To further analyze the mechanisms behind this break of specific immune tolerance, we characterized rFVIIa-specific T cells by the expression of CD154, a marker of antigen-specific T cells. Cytokine production and CD154 expression were assessed upon re-stimulation with rFVIIa. In contrast to mice that were immunized with rFVIIa only, we found increased numbers of rFVIIa-specific T cells in rFVIIa-LPS-treated mice displaying a stable, highly pro-inflammatory (IL-2+/IFN-g+) memory phenotype. These data could suggest that rFVIIa-specific Teff that escaped clonal deletion during induction of central immune tolerance, are present in the periphery of human FVII-transgenic mice. This would imply that rFVIIa-specific Teff could be actively suppressed by Tregs. This suppression could be overcome by danger signals like LPS. We currently study the regulatory mechanisms that maintain tolerance upon administration of FVIIa without LPS. We are approaching this question by correlating the characteristics of FVIIa-specific Teff and Treg responses under both tolerant and non-tolerant conditions. Ultimately, we aim to understand which danger signals have to be provided to break immune tolerance and how tolerance is regulated. Understanding these regulatory mechanisms will enable us to develop new therapeutic strategies and prevent conditions that lead to the induction of antibodies against drug products in patients. Disclosures: Lenk: Baxter BioScience: Employment. Pasztorek:Baxter BioScience: Employment. Weiller:Baxter BioScience: Employment. Ahmad:Baxter BioScience: Employment. Schwarz:Baxter BioScience: Employment. Scheiflinger:Baxter BioScience: Employment. Reipert:Baxter Innovations GmbH: Employment. de la Rosa:Baxter BioScience: Employment.


Blood ◽  
2013 ◽  
Vol 121 (22) ◽  
pp. 4449-4456 ◽  
Author(s):  
David W. Scott ◽  
Kathleen P. Pratt ◽  
Carol H. Miao

Abstract A major problem in treating hemophilia A patients with therapeutic factor VIII (FVIII) is that 20% to 30% of these patients produce neutralizing anti-FVIII antibodies. These antibodies block (inhibit) the procoagulant function of FVIII and thus are termed “inhibitors.” The currently accepted clinical method to attempt to eliminate inhibitors is immune tolerance induction (ITI) via a protocol requiring intensive FVIII treatment until inhibitor titers drop. Although often successful, ITI is extremely costly and is less likely to succeed in patients with high-titer inhibitors. During the past decade, significant progress has been made in clarifying mechanisms of allo- and autoimmune responses to FVIII and in suppression of these responses. Animal model studies are suggesting novel, less costly methods to induce tolerance to FVIII. Complementary studies of anti-FVIII T-cell responses using blood samples from human donors are identifying immunodominant T-cell epitopes in FVIII and possible targets for tolerogenic efforts. Mechanistic experiments using human T-cell clones and lines are providing a clinically relevant counterpoint to the animal model studies. This review highlights recent progress toward the related goals of lowering the incidence of anti-FVIII immune responses and promoting durable, functional immune tolerance to FVIII in patients with an existing inhibitor.


1995 ◽  
Vol 4 (4) ◽  
pp. 361-370 ◽  
Author(s):  
Ronald G. Gill ◽  
Leslie Wolf

The goal of cellular transplantation is to allow long-term function of the grafted cells using minimal host immunosuppression. To this end, the major strategies to implant cells and tissues are through: (i) the pretreatment of the graft to reduce tissue immunogenicity; (ii) the application of immunoisolation technologies to prevent host sensitization to implanted cells; and (iii) the induction of immunological tolerance to the donor tissues. Further, a major dilemma facing clinical tissue grafting is the shortage of donor tissue for transplantation. This problem requires the consideration of tissues from other species (xenografts) as a potential source of donor material. In light of these issues, the focus of this discussion is on the T cell-dependent response to allogeneic and xenogeneic transplants and the implications of this reactivity on the field of cellular replacement therapy.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1029-1029
Author(s):  
Marc G. Jacquemin ◽  
Renaud Lavend’homme ◽  
Thérèse Tinot ◽  
Kathelijne Peerlinck ◽  
Jean Marie Saint-Remy ◽  
...  

Abstract In patients with hemophilia A and FVIII inhibitor, tolerance to FVIII can frequently be restored by prolonged administration of FVIII (immune tolerance induction). We studied the cellular response to FVIII in two patients successfully desensitized with high doses of FVIII. Each patient had also transiently received glucocorticoids, when the antibody titer increased after an initial decline upon start of the desensitization. On repeated stimulations with FVIII loaded autologous dendritic cells, FVIII-specific T oligoclonal cell lines and T cell clones were derived from one of the two patients up to two years after complete elimination of the inhibitor. The interleukins produced by the T cell lines included IL-2, IL-5 and IL-13. Although ITI is frequently unsuccessful in mild/moderate haemophilia A patients, this patient remained tolerant to FVIII even after a 10 days FVIII continuous infusion, 18 months after complete elimination of the inhibitor. Several microcultures from CD4+ T cells purified after this treatment responded to FVIII specifically demonstrating the long term persistence of FVIII-specific T cells after ITI. By contrast, no FVIII-specific T cell lines could be derived from blood of control individuals without hemophilia A. Thus, although in animal models of tolerance, administration of high doses of antigen result in T cell elimination, deletion of FVIII-specific T cells is not required for successful tolerance induction to FVIII.


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.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2209-2209
Author(s):  
Debalina Sarkar ◽  
Gongxian Liao ◽  
Cox Terhorst ◽  
Roland W Herzog

Abstract Abstract 2209 In vivo induction and expansion of Treg is a powerful tool to limit unwanted immune responses and promote tolerance. For example, we have been successful inducing tolerance to factors VIII and FIX in hemophilic mice when the coagulation factor antigen was administered with the mTOR inhibitor rapamycin (J Thromb Haemost 7:1523 and 9:1524, Front Microbiol 2:244). Rapamycin, a macrocyclic triene antibiotic, is an immunosuppressant used to avoid transplant rejection. It suppresses the mTOR1 (and upon prolonged exposure also mTOR2) signaling pathway. Importantly, while mTOR blockage results in deletion of Teff, Treg can be induced and expanded because they are able to utilize alternative (STAT) signaling pathways. Others have shown that existing Treg can be expanded in vivo upon administration of Fms-like tyrosine kinase ligand-3 (Flt3L), a cytokine that drives generation of dendritic cells (DC) from hematopoietic progenitor cells and DC proliferation. This link between DC homeostasis and Treg is evident from the low Treg numbers found in Flt3L-deficient mice and from prevention of graft vs host disease upon treatment with Flt3L. This raises the question of whether a combined approach of rapamycin administration and Flt3L-induced DC generation would result in an optimal immune tolerance protocol. Interestingly, it has been reported that rapamycin blocks Flt3L-induced differentiation of progenitor cells into DC, indicating that Flt3L signaling in DC occurs through the mTOR pathway. However, we find in mice transgenic for an ovalbumin-specific CD4+ T cell receptor (but deficient in recombinase activating gene, rag-2) that ova peptide antigen administration results in substantially enhanced deletion of Teff and in induction of CD4+CD25+FoxP3+CD62L+GITR+ Treg when combined with these two drugs. This was accomplished by repeated administration (twice per week) of a cocktail of the 3 components. Antigen plus either drug causes Teff deletion, while rapamycin is required for Treg induction (which is further enhanced by Flt3L). Antigen, rapamycin, and Flt3L all impact changes in the numbers and frequencies of DC subsets in the spleen during the regimen. The combination all 3 components most potently directs a substantial (3–5 fold, P<0.001) increase in CD11cloPDCA+ plasmacytoid DC numbers (but not of conventional CD11chiPDCA− DCs). While pDCs are known to provide innate anti-viral responses, they also play an important role in immune tolerance. Consequently, when pDC were partially depleted with anti-PDCA, Treg induction was significantly impaired. Furthermore, the protocol caused an increase in the frequency of Indoleamine-pyrrole 2,3-dioxygenase (IDO)-expressing pDCs (which is known to activate resting Treg for suppressor activity). Finally, FLt3L-induced expansion of Treg (but not of DCs) is less effective in GITR-L −/− mice. Combined, these data demonstrate that i) Flt3L and rapamycin can be used synergistically for induction of T cell tolerance, ii) pDCs can be expanded within a rapamycin regimen, iii) and Fl3tL-induced pDC expansion facilitates Treg induction, which is partially dependent on GITR-L (a co-stimulatory molecule primarily expressed by pDCs that promotes cross talk to Treg by engagement of the GITR receptor). In order to establish relevance of this protocol for treatment of disease, we intravenously injected a F.VIII protein/rapamycin/Flt3L cocktail into hemophilia A mice (C57BL6/129 F8e16 −/−) twice per week for 1 month. Subsequently, mice received 1 month of factor replacement therapy (1 IU human FVIII, IV, once per week). Control mice without prior immune modulatory regiment or that received non-specific immune suppression (rapamycin and Flt3L only) formed high-titer inhibitors against FVIII (70–80 BU), which was significantly suppressed to ∼10 BU (P<0.001, n=5 per group). Importantly, inhibitor titers were only mildly reduced (to ∼40 BU) when Flt3L was omitted from the tolerogenic cocktail, thereby confirming the synergistic effect of flt3L and rapamycin in tolerance induction. This approach combines expansion of regulatory antigen presenting and T cells and should be of broad relevance for cell and organ transplantation as well as for treatment of inherited protein deficiencies and of autoimmune diseases. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document