scholarly journals Major role of local immune responses in antibody formation to factor IX in AAV gene transfer

Gene Therapy ◽  
2005 ◽  
Vol 12 (19) ◽  
pp. 1453-1464 ◽  
Author(s):  
L Wang ◽  
O Cao ◽  
B Swalm ◽  
E Dobrzynski ◽  
F Mingozzi ◽  
...  
Blood ◽  
2007 ◽  
Vol 110 (4) ◽  
pp. 1132-1140 ◽  
Author(s):  
Ou Cao ◽  
Eric Dobrzynski ◽  
Lixin Wang ◽  
Sushrusha Nayak ◽  
Bethany Mingle ◽  
...  

Abstract Gene replacement therapy is complicated by the risk of an immune response against the therapeutic transgene product, which in part is determined by the route of vector administration. Our previous studies demonstrated induction of immune tolerance to coagulation factor IX (FIX) by hepatic adeno-associated viral (AAV) gene transfer. Using a regulatory T-cell (Treg)–deficient model (Rag-2−/− mice transgenic for ovalbumin-specific T-cell receptor DO11.10), we provide first definitive evidence for induction of transgene product-specific CD4+CD25+ Tregs by in vivo gene transfer. Hepatic gene transfer–induced Tregs express FoxP3, GITR, and CTLA4, and suppress CD4+CD25− T cells. Tregs are detected as early as 2 weeks after gene transfer, and increase in frequency in thymus and secondary lymphoid organs during the following 2 months. Similarly, adoptive lymphocyte transfers from mice tolerized to human FIX by hepatic AAV gene transfer indicate induction of CD4+CD25+GITR+ that suppresses antibody formation to FIX. Moreover, in vivo depletion of CD4+CD25+ Tregs leads to antibody formation to the FIX transgene product after hepatic gene transfer, which strongly suggests that these regulatory cells are required for tolerance induction. Our study reveals a crucial role of CD4+CD25+ Tregs in preventing immune responses to the transgene product in gene transfer.


Blood ◽  
2004 ◽  
Vol 103 (1) ◽  
pp. 143-151 ◽  
Author(s):  
Jun Zhang ◽  
Lingfei Xu ◽  
Mark E. Haskins ◽  
Katherine Parker Ponder

Abstract The effect of neonatal gene transfer on antibody formation was determined using a retroviral vector (RV) expressing human factor IX (hFIX). Normal mice from different strains injected intravenously with RV as newborns achieved therapeutic levels of hFIX without antibody production and were tolerant as adults to challenge with hFIX. Neonatal hemophilia B mice that received different amounts of RV achieved stable and dose-related expression of hFIX without anti-hFIX antibody formation. After protein challenge, antibody formation was markedly reduced for animals that expressed hFIX at levels higher than 14 ng/mL (0.3% of normal). However, antibodies developed for animals that received the lowest dose of RV and expressed hFIX at approximately 2 ng/mL before protein challenge. In dogs, neonatal injection of a high dose of RV resulted in 500 ng/mL hFIX in plasma without antibody formation. We conclude that neonatal gene transfer with RV does not induce antibody responses to hFIX in mice or dogs and that mice achieving levels greater than 3 × 10–10 M hFIX are usually tolerant to protein injection as adults. Low-dose gene therapy or frequent protein injections in the neonatal period might induce tolerance to subsequent injections of protein with a low risk for adverse effects.


2000 ◽  
Vol 1 (3) ◽  
pp. 225-235 ◽  
Author(s):  
Paul A. Fields ◽  
Dariusz W. Kowalczyk ◽  
Valder R. Arruda ◽  
Elina Armstrong ◽  
Mark L. McCleland ◽  
...  

2014 ◽  
Vol 12 (1) ◽  
pp. 25 ◽  
Author(s):  
Geoffrey L Rogers ◽  
Ashley T Martino ◽  
Irene Zolotukhin ◽  
Hildegund CJ Ertl ◽  
Roland W Herzog

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3185-3185
Author(s):  
E. Dobrzynski ◽  
F. Mingozzi ◽  
L. Wang ◽  
B. Mingle ◽  
O. Cao ◽  
...  

Abstract The use of gene replacement therapy is an attractive approach for the treatment of the genetic bleeding disorder hemophilia B (caused by mutations in the coagulation factor IX, FIX, gene). A major concern with this type of procedure is the potential for a host immune response to the therapeutic gene product, which would render treatment ineffective. Previously, we observed inflammatory, cytotoxic T lymphocyte, and antibody responses to a human FIX (hFIX) transgene product after intramuscular (IM) delivery via an E1/E3-deleted adenoviral vector (Ad-hFIX) in C57BL/6 mice. Different from this Th1-biased immune response, IM injection of adeno-associated viral (AAV) vector, a Th2-biased, non-inflammatory response led to antibody-mediated neutralization of hFIX expression, without CTL activation. In contrast to these observations on muscle-directed vector administration, hepatic AAV-hFIX gene transfer induced immune tolerance to the transgene product (JCI 111:1347). Lack of anti-hFIX formation was demonstrated even after challenge with hFIX in adjuvant. In order to examine the effect of tolerance induction on CD8+ T cell-mediated cellular immune responses, we performed the following experiments. C57BL/6 mice (n=4 per experimental group) received IM injections of AAV-hFIX vector (serotype 1) in one hind limb and/or Ad-hFIX vector in the contra-lateral leg. In the latter case, inflammation (as determined by H&E histological evaluation), CD8+ T cell infiltrate and destruction of hFIX expressing muscle fibers were obvious in both legs because of the Ad-hFIX mediated activation of CTL to hFIX. CD8+ T cell responses were strongest in Ad-hFIX transduced muscle at day 14 and in the AAV-hFIX leg at day 30. Expression of hFIX as determined by immunohistochemistry became undetectable in Ad-hFIX injected muscle by day 30, but was not completely eliminated in AAV-hFIX transduced muscle. Injection of AAV-hFIX only, did not cause inflammation of muscle tissue or CD8+ cell infiltrate. When the identical experiment was carried out in C57BL/6 mice that were expressing hFIX from hepatic gene transfer via the AAV serotype 2 vector (performed 6 weeks earlier), a substantial increase in systemic hFIX expression was observed after IM administration of the Ad and AAV-1 vectors (again injected into contra-lateral legs). However, a portion of the increased expression was subsequently lost, which correlated with inflammation and CD8+ T cell infiltrate of the Ad-hFIX transduced muscle. Interestingly, no (3/4 mice) or only minor (1/4 mice) infiltrate was observed in AAV-hFIX injected muscles. Consequently, hFIX expression persisted in the AAV, but not the Ad transduced legs. Presumably, CTL responses to adenoviral antigens were sufficient to target Ad-hFIX transduced muscle despite tolerance to the transgene product. In contrast to control mice, hepatic tolerized animals failed to form anti-hFIX after challenge by IM injection of these viral vectors. Moreover, inflammatory and destructive cellular immune responses to the transgene product were successfully prevented by hepatic tolerance induction, indicating that tolerance induced by gene transfer to the liver affects cellular as well as antibody-mediated responses and extents to tissues other than liver.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 452-452
Author(s):  
Ou Cao ◽  
Lixin Wang ◽  
Sushrusha Nayak ◽  
Roland W. Herzog

Abstract Gene therapy for the X-linked bleeding disorder hemophilia B may be limited by immune responses to the factor IX (F.IX) gene product. Hepatic adeno-associated virus (AAV) gene transfer can induce immune tolerance to F.IX (JCI111:1347, PNAS103:4592). Tolerance is associated with activation of regulatory cells that suppress antibody formation to F.IX. In order to identify these regulatory cells, splenocytes of C57BL/6 mice tolerized to human F.IX (hF.IX) by heptic gene transfer (portal vein injection of 1x1011 AAV vector genomes) were adoptively transferred to naive mice of the same strain. Recipient mice were immunized with hF.IX in adjuvant on the next day. Compared to cells transferred from control animals (no gene transfer), total splenocytes, CD4+ cells, or CD4+CD25+ cells were equally efficient in suppression of anti-hF.IX formation (n=7–8 per experimental group, P<0.02 for comparison to total splenocytes, CD4+ cells, or CD4+CD25- cells of controls), while CD4- cells failed to suppress, and CD4+CD25- cells were inefficient. CD4+CD25+ from naive control mice, which contain regulatory T cells but lack specificity for hF.IX, gave highly variable results and on average failed to suppress. When tolerized C57BL/6 mice were challenged with hF.IX/adjuvant, the animals lacked antibody formation to hF.IX and in vitro cytokine release and showed an ~2-fold increase in FoxP3 message in splenic CD4+ cells in vivo. Taken together, these data indicate that induction of regulatory CD4+CD25+ T cells is part of the tolerance mechanism. However, the significance of this finding was unclear. In the next experiment, C57BL/6 mice received hepatic AAV-hF.IX gene transfer and were additionally injected with rat anti-mouse CD25 or with isotype control rat IgG (ip injections at days 0, 14, 28, and 42, n=5 per group). Analysis of peripheral blood cells by flow cytometry showed presence of CD4+CD25+ cells at a frequency of 8–10% in controls and undetectable levels in anti-CD25 treated mice. By day 49, 4/5 anti-CD25 treated mice had a low-titer, but detectable antibody (IgG1) to hF.IX. Subsequent challenge with hF.IX/cF.IX caused a rise in anti-hF.IX to 0.5–2 μg/ml in 3/5 anti-CD25 treated mice within 3 weeks. None of the mice treated with control IgG (0/5) developed a detectable antibody to hF.IX even after challenge. These data demonstrate that CD4+CD25+ regulatory T cells are required for tolerance induction to F.IX. Thus far, we failed to break tolerance by depletion of CD25+ cells at later time points (i.e. during the maintenance phase of tolerance, when other mechanisms such as T cell anergy and deletion may become more prevalent). To obtain definitive evidence for induction of CD4+CD25+ Treg, hepatic AAV-ova gene transfer was performed in DO11.10-tg Rag-2 −/− BALB/c mice, which are deficient in Treg. The DO11.10 T cell receptor is specific for ova peptide 323–339/MHC class II I-Ad complex. Within 2 weeks after gene transfer, CD4+CD25+GITR+ cells emerged in the thymus and in secondary lymphoid organs. Frequency of these cells increased to 2–4% by 2 months and subsequently remained at that level. These cells also expressed CTLA-4 and FoxP3 (>100-fold increase in FoxP3 message compared to CD4+ cells from naive mice or compared to CD4+CD25- cells of AAV-ova transduced mice), and efficiently suppressed CD4+CD25- cells in vitro. In summary, hepatic AAV gene transfer induces transgene product-specific CD4+CD25+ Treg, which suppress antibody formation to the transgene product and are required for tolerance induction. These results should have broad implications for in vivo gene transfer.


2011 ◽  
Vol 19 (7) ◽  
pp. 1263-1272 ◽  
Author(s):  
Brad E Hoffman ◽  
Ashley T Martino ◽  
Brandon K Sack ◽  
Ou Cao ◽  
Gongxian Liao ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4209-4209
Author(s):  
Geoffrey L. Rogers ◽  
Roland W Herzog

Abstract Self-complementary adeno-associated viral (scAAV) vectors have become a popular tool for AAV gene therapies due to their increased transgene expression relative to single-stranded (ss)AAV vectors, at least for models in which the transgene can fit within these restrictive constructs. The most recent clinical trial for hemophilia B used a scAAV8 vector. However, recent reports by our lab and others have suggested that these vectors are also more immunogenic than their single-stranded brethren. We previously showed that, during hepatic gene transfer, the innate immune response is greater with a scAAV vector. Moreover, Wu et al. demonstrated an enhanced T and B cell response to a secreted form of HIV’s gag protein when it was delivered via scAAV. While this was beneficial for their vaccine model, it could potentially be problematic for therapeutic gene transfer applications. To investigate whether this phenomenon could deleteriously impact gene therapy for hemophilia B, we used a partial knockout model of hemophilia B. This mouse model has the murine factor IX gene knocked out, and then a truncated form of human factor IX (hF.IX) is inserted. Specifically, the mice we used have a late stop codon (LS) at amino acid 338; while hF.IX can be detected in the liver, this mutated protein is not released into circulation (crm-). During muscle gene transfer with AAV2, LS mice have an interesting response: while they form antibodies to hF.IX, there is no CD8+ T cell infiltrate detected in the transduced tissue. We then set out to determine whether this partial tolerance could be disrupted using a scAAV vector. Using AAV1, which is a more relevant serotype for muscle gene therapy, we injected single-stranded and self-complementary vectors (1011 vg/mouse) and measured the resulting immune responses. Surprisingly, unlike with AAV2, the LS mice completely tolerated AAV1-mediated intramuscular delivery of hF.IX. They did not make detectable anti-hF.IX IgG1, and their plasma had no Bethesda activity through 4 weeks post-injection. In contrast, hF.IX null mice (HBKO) made 10708±3869 ng anti-hF.IX IgG1 and developed inhibitory activity of 22.5±26 Bethesda units 4 weeks after delivery of a scAAV vector. Consequently, LS mice had circulating hF.IX levels of 148.9±18.64 (ssAAV1) and 60.12±8.71 ng/mL (scAAV1), while hF.IX was undetectable in the plasma of HBKO mice at this time point. The CD8 response was similarly lacking in LS mice, with splenic responses to hF.IX detected by ELISPOT reduced by ∼2-10 fold relative to HBKO mice. Infiltrating CD8 cells were detected in the muscle of HBKO but not LS mice. In conclusion, our data suggest that the underlying mutation within the patient is a more important risk factor for immune responses to the transgene than the molecular form of the AAV genome. Disclosures: Herzog: Genzyme: AAV technology, AAV technology Patents & Royalties.


Sign in / Sign up

Export Citation Format

Share Document