scholarly journals 489. Humoral Tolerance to FVIII in Hemophilia A Mice Using Cytokine-Conditioned Dendritic Cells

2012 ◽  
Vol 20 ◽  
pp. S190
Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 213-213 ◽  
Author(s):  
Margaret V. Ragni ◽  
Wenhu Wu ◽  
Xiaoyan Liang ◽  
Lina Lu

Abstract Inhibitor formation is a severe complication of hemophilia, occurring in up to 25% and associated with poor response to factor replacement, uncontrolled bleeding, and high morbidity. Preventing inhibitor formation is, thus, a major goal of hemophilia management. The role of dendritic cells (DC) in regulating immune response has been increasingly recognized: immature DC (imDC) induce T regulatory cells in vitro and promote Ag-specific tolerance in vivo. We, therefore, studied the role of imDC propagated from bone marrow with GM-CSF + TGFβ to prevent inhibitor formation in the hemophilia A murine model. Following tail vein injection of recombinant F.VIII (Advate, Baxter) 2.5 U (0.2 μg) on days 0, 2, and 4 in hemophilia A exon 16 KO C57Bl/6 mice, anti-VIII antibodies were detected by semi-quantitative APTT (scored 1-4), peaking on day 6. On rechallenge with F.VIII 2.5 U on days 12, 14, and 16, anti-VIII was detected, peaking on day 17. Anti-VIII production was associated with high level splenic T cell proliferation in response to F.VIII stimulation in vitro, measured by 3H-thymidine incorporation in mixed lymphocyte reaction (MLR). By contrast, there was no antibody formation in F.VIII-treated Wt C57Bl/6 mice: the latter was associated with low T cell response to F.VIII in vitro. Functionally immature DC (imDC) were propagated from the bone marrow of hemophilia A mice with GM-CSF (4ng/ml) and TGFβ (0.2ng/ml). For comparison, functionally mature dendritic cells (mDC) were propagated with GM-CSF (4ng/ml) and IL-4 (1000U/ml).The former (imDC) demonstrated deficient NF-kB binding activity in nuclear protein as detected by gel shifting assay and expressed low level of costimulatory molecules CD80, CD86; by contrast, the latter (mDC) demonstrated enhanced NF-kB binding activity and high levels of co-stimulatory molecules. Administration of 2x106 F.VIII-pulsed imDC (20U/ml x 24h) 7 days before F.VIII dosing on days 0, 2, and 4, led to reduction in inhibitor formation on day 6 (score 1.6 vs. 2.3 in control group) which was further reduced on day 8 (score 1.0 vs. 2.0 in control group). The inhibitor could not be detected on day 8 in 2 of 4 mice pretreated with F.VIII-pulsed imDC. By contrast, high levels of inhibitor were detected in mice pretreated with F.VIII-pulsed mDC (score 3.3). Rechallenge with F.VIII on day 10 in imDC-treated mice resulted in no increase in the reduced or absent anti-VIII effect on day 12. Splenic T cells (CD3+) from the imDC-pretreated mice showed lower proliferative capacity when restimulated in vitro with F.VIII, suggesting that imDC induced F.VIII unresponsiveness. These studies show that FVIII-pulsed imDC reduce the intensity of inhibitor formation, and suggest the potential role of modified DC in preventing or reducing F.VIII inhibitor formation.


2009 ◽  
Vol 37 (6) ◽  
pp. 744-754 ◽  
Author(s):  
Margaret V. Ragni ◽  
Wenhu Wu ◽  
Xiaoyan Liang ◽  
Ching-Chuan Hsieh ◽  
Andrea Cortese-Hassett ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2387-2387
Author(s):  
Glaivy Batsuli ◽  
Seema R Patel ◽  
Courtney Cox ◽  
Wallace H. Baldwin ◽  
John S. Lollar ◽  
...  

Introduction: The immune response to factor VIII (fVIII) is a CD4+ T cell dependent process initiated by fVIII recognition and processing by antigen presenting cells. The C1 and C2 domains have been proposed as the primary domains that mediate fVIII internalization by dendritic cells. Our prior studies demonstrate that fVIII pre-bound to anti-C1 and C2 domain monoclonal antibodies (MAbs) reduces fVIII internalization by murine derived dendritic cells. However, anti-A1 and A3 domain antibodies increase fVIII endocytosis by dendritic cells. In this study, we analyzed the antibody titers of hemophilia A mice immunized with binary fVIII and MAb complexes to determine whether changes in fVIII internalization observed in vitro correspond to changes in the immune response to fVIII in vivo. Methods: Exon16 (E16) knockout mice deficient in fVIII were immunized with B-domain deleted fVIII in the presence of either anti-fVIII MAb 2-116 (anti-A1, IgG2a), 4A4 (anti-A2, IgG2a), 2-113 (anti-A3, IgG1), B136 (anti-C1, IgG2a), or 3D12 (anti-C2, IgG2b) versus fVIII alone. Mice were immunized by weekly retro-orbital injections of 0.1 µg fVIII incubated with 1 µg anti-fVIII MAb for 4 weeks followed by a boost dose of 0.2 µg fVIII and 2 µg anti-fVIII MAb one week later at week 5. Anti-fVIII ELISA titers (in arbitrary units, AU) were analyzed from plasma samples collected at week 7 to account for IgG half-life. A separate cohort of mice immunized with anti-fVIII MAbs alone served as controls to normalize ELISA titers in each fVIII/MAb group to account for residual injected MAbs in the plasma samples. Results: Hemophilia A mice immunized with fVIII/2-116 and fVIII/4A4 complexes significantly increased anti-fVIII ELISA titers compared to mice immunized with fVIII alone (figure 1). Mice immunized with fVIII/2-116 and fVIII/4A4 had median ELISA titers of 7,401 AU (interquartile range, IQR, 2,654 - 11,910 AU) and 3,620 AU (IQR 1,062 - 7,969 AU), respectively, compared to mice immunized with fVIII (median titer 1,063 AU, IQR 402 - 2,476 AU). MAb 2-116 is a non-inhibitory antibody with a titer of <1 Bethesda Unit (BU)/mg IgG, while 4A4 is a highly inhibitory antibody with a titer of 40,000 BU/mg IgG. Neither MAb interferes with fVIII binding to von Willebrand factor (VWF) or phospholipid vesicles. Mice immunized with fVIII/2-113 (median titer 2,210 AU, IQR 402 - 8,318 AU), fVIII/B136 (median titer 123 AU, IQR 0 - 9,709 AU), and fVIII/3D12 (median titer 3,244 AU, IQR 0 - 5,180 AU) did not have significantly different anti-fVIII ELISA titers compared to mice immunized with fVIII. However there was a trend towards reduced anti-fVIII titers with fVIII/B136 and fVIII/3D12 injections. MAbs B136 and 3D12 inhibit fVIII binding to VWF and have been shown to significantly increased fVIII clearance in hemophilia A mice compared to fVIII in a VWF-dependent manner. Median titers for mice immunized with MAbs alone to account for residual injected MAbs were 13 AU (2-116), 12 AU (4A4), 18 AU (2-113), 16 AU (B136), and 4 AU (3D12). Conclusions: Immunization of hemophilia A mice with fVIII/MAb complexes, specifically anti-A1 MAb 2-116 and anti-A2 MAb 4A4, enhance the immune response to fVIII. MAb 2-116 significantly increased anti-fVIII antibody titers in vivo, which correlates with increased fVIII internalization by immature dendritic cells observed in vitro. A better understanding of the effect of anti-fVIII antibodies on fVIII conformational changes could provide insight into whether these changes alter fVIII recognition by immune cells and subsequently propagate the immune response to fVIII at the onset of inhibitor formation or during immune tolerance induction. Disclosures Batsuli: Genentech: Other: Advisory board participant; Bayer: Other: Advisory board participant; Octapharma: Other: Advisory board participant. Meeks:HEMA Biologics: Membership on an entity's Board of Directors or advisory committees; Novo Nordisk: Membership on an entity's Board of Directors or advisory committees; Genentech: Membership on an entity's Board of Directors or advisory committees; Bioverativ: Membership on an entity's Board of Directors or advisory committees; Takeda-Shire: Membership on an entity's Board of Directors or advisory committees; Bayer: Membership on an entity's Board of Directors or advisory committees.


2012 ◽  
Vol 23 (7) ◽  
pp. 769-780 ◽  
Author(s):  
Gautam Sule ◽  
Masataka Suzuki ◽  
Kilian Guse ◽  
Racel Cela ◽  
John R. Rodgers ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 199-199
Author(s):  
Aleksandra Wroblewska ◽  
Simon D van Haren ◽  
Eszter Herczenik ◽  
Aleksandra Rusiecka ◽  
Paul H Kaijen ◽  
...  

Abstract Abstract 199FN2 The X-chromosome-linked bleeding disorder hemophilia A is caused by the absence or dysfunction of blood coagulation factor VIII (FVIII) which can be corrected by regular intravenous administration of FVIII. Development of antibodies directed against FVIII (referred to as FVIII inhibitors) is common complication in hemophilia care. The formation of FVIII-neutralizing antibodies in hemophilia A patients is initiated by the endocytosis of FVIII by professional antigen-presenting cells. Endocytosis of FVIII by human monocyte-derived dendritic cells can be significantly blocked by monoclonal antibody KM33, directed towards the C1 domain of FVIII. We have previously shown that C1 domain residues 2092 and 2093 are required for KM33 binding to FVIII (Meems et al., 2009). However, replacement of both these residues by Ala did not completely abolish KM33 binding. We created an additional C1 domain variant which included an arginine at position 2090. A FVIII variant in which three residues, 2090, 2092 and 2093 were substituted by alanine, showed only minimal binding to KM33. Functional analysis revealed that this variant designated FVIII-2090/2092/2093 retains pro-coagulant activity as measured by a chromogenic assay. FVIII-2090/2092/2093 displayed a strongly reduced internalization by human monocyte-derived dendritic cells and macrophages, as well as murine bone marrow-derived dendritic cells. These results emphasize a role for C1 domain residues 2090, 2092 and 2093 in FVIII endocytosis by antigen-presenting cells. We subsequently investigated the in vivo ability of this variant to induce inhibitors in FVIII−/− mice (E17KO). We show that E17KO mice treated with the FVIII variant have significantly lower anti-FVIII antibody titers when compared to mice treated with wild type FVIII. In accordance with these findings reduced numbers of FVIII-specific antibody-secreting cells were detected in the spleen of mice treated with FVIII-2090/2092/2093. Also, FVIII-specific CD4+ T cell responses of splenocytes derived from FVIII-2090/2092/2093 infused mice were greatly reduced when compared to that of splenocytes derived from wild type FVIII infused mice. These findings show that alanine substitutions at positions 2090, 2092 and 2093 result in a FVIII molecule that is significantly less immunogenic when compared to wild type FVIII. Collectively, our data suggest that FVIII variants displaying a reduced uptake by antigen-presenting cells show a reduced immunogenicity in vivo. Based on our findings we hypothesize that FVIII variants displaying a reduced uptake by antigen-presenting cells provide a novel therapeutic approach to reduce inhibitor development in patients with hemophilia A. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 119 (22) ◽  
pp. 5294-5300 ◽  
Author(s):  
Aleksandra Wroblewska ◽  
Simon D. van Haren ◽  
Eszter Herczenik ◽  
Paul Kaijen ◽  
Aleksandra Ruminska ◽  
...  

Abstract Development of neutralizing Abs to blood coagulation factor VIII (FVIII) provides a major complication in hemophilia care. In this study we explored whether modulation of the uptake of FVIII by APCs can reduce its intrinsic immunogenicity. Endocytosis of FVIII by professional APCs is significantly blocked by mAb KM33, directed toward the C1 domain of FVIII. We created a C1 domain variant (FVIII-R2090A/K2092A/F2093A), which showed only minimal binding to KM33 and retained its activity as measured by chromogenic assay. FVIII-R2090A/K2092A/F2093A displayed a strongly reduced internalization by human monocyte-derived dendritic cells and macrophages, as well as murine BM-derived dendritic cells. We subsequently investigated the ability of this variant to induce an immune response in FVIII-deficient mice. We show that mice treated with FVIII-R2090A/K2092A/F2093A have significantly lower anti-FVIII Ab titers and FVIII-specific CD4+ T-cell responses compared with mice treated with wild-type FVIII. These data show that alanine substitutions at positions 2090, 2092, and 2093 reduce the immunogenicity of FVIII. According to our findings we hypothesize that FVIII variants displaying a reduced uptake by APCs provide a novel therapeutic approach to reduce inhibitor development in hemophilia A.


2020 ◽  
Vol 18 (1) ◽  
pp. 201-216
Author(s):  
Vincent P. Diego ◽  
Bernadette W. Luu ◽  
Marco Hofmann ◽  
Long V. Dinh ◽  
Marcio Almeida ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document