Gene Transfer and Tolerance Induction

Author(s):  
W. Wong ◽  
J. Fry ◽  
K. Hyde ◽  
P. Morris ◽  
K. Wood
Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2154-2154
Author(s):  
Luo Xiaofeng ◽  
Jocelyn A. Schroeder ◽  
Christina Baumgartner ◽  
Juan Chen ◽  
Jianda Hu ◽  
...  

Abstract Induction of antigen-specific immune tolerance is desirable in autoimmune diseases, transplantation, and gene therapy. Our previous studies have demonstrated that FVIII or FIX expression ectopically targeted to platelets under control of the platelet-specific αIIb promoter results in transgene protein storage in platelet α-granules. Further studies have demonstrated that lentivirus-mediated platelet-specific gene delivery to hematopoietic stem cells (HSCs) not only restores hemostasis but also induces antigen-specific immune tolerance in hemophilic mice. We wanted to explore whether platelet-specific gene transfer can be used as a means of immune tolerance induction. In the current study, we used ovalbumin (OVA) as a non-coagulant protein to further examine the potential of a platelet gene therapy-based immune tolerance induction approach. We constructed a lentiviral vector (LV) in which OVA is driven by the αIIb promoter (2bOVA). Evidence suggests that VWF propeptide can reroute unrelated secreting proteins to a storage pathway. Thus, we designed another vector, 2bVpOVA, which contains VWF propeptide to secure OVA storage in platelet granules. HSCs from wild type B6/CD45.2 mice were transduced with 2bOVA or 2bVpOVA LV and transplanted into B6/CD45.1 recipients preconditioned with 660 cGy total body irradiation. We found that 96% of OVA expression in whole blood was stored in platelets with a level of 51.3 ± 22.5 ng/108 platelets (n = 5) while 4% was detectable in plasma in 2bOVA-transduced recipients at 12-week after transplantation. This distribution is very similar to the results we obtained from the FIX study. In contrast, 98% of OVA was stored in platelets with a level of 3.9 ± 3.3 ng/108 platelets (n = 5) in 2bVpOVA-transduced recipients. The lower total OVA expression level in the 2bVpOVA group could be due to the size effect of transgene expression cassette as the 2bVpOVA cassette is 3-fold larger than the 2bOVA cassette. To investigate whether anti-OVA immune tolerance was established in recipients after platelet-specific OVA gene transfer, 16-weeks post-transplantation, animals were challenged with OVA. The titer of anti-OVA total IgG determined by ELISA assay was 640 ± 101 in the 2bOVA group and 320 ± 0 in the 2bVpOVA group. These titers were significantly lower than that obtained from the untransduced control group (10210 ± 3636), demonstrating that platelet-specific OVA gene delivery to HSCs can suppress the anti-OVA immune response. Of note, the titer of anti-OVA total IgG in the 2bVpOVA group was significantly lower than in the 2bOVA group although the total OVA expression levels in the 2bOVA group is 13-fold higher than in the 2bVpOVA group. The percentage of regulatory T cells in peripheral blood in 2bOVA and 2bVpOVA-transduced recipients was significantly higher than in untransduced control animals. In summary, our data demonstrate that targeting transgene expression and storage in platelet a-granules is a potentially promising approach for inducing immune tolerance. Disclosures No relevant conflicts of interest to declare.


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.


2005 ◽  
Vol 3 (4) ◽  
pp. 234-240 ◽  
Author(s):  
E. Dobrzynski ◽  
R. W. Herzog

Hematology ◽  
2003 ◽  
Vol 2003 (1) ◽  
pp. 559-574 ◽  
Author(s):  
Margaret E. Rick ◽  
Christopher E. Walsh ◽  
Nigel S. Key

Abstract Both clinical and basic problems related to the congenital bleeding disorders continue to confront hematologists. On the forefront are efforts to bring genetic correction of the more common bleeding disorders such as hemophilia A to the clinic in a safe and accessible manner. A second issue, particularly for patients with hemophilia, is the development of inhibitors—questions of how they arise and how to prevent and treat these problems that confound otherwise very successful replacement therapy and allow patients to maintain normal lifestyles. A third issue is the continuing question of diagnosis and management of von Willebrand disease, the most common congenital bleeding disorder, especially in individuals who have borderline laboratory values, but have a history of clinical bleeding. In Section I, Dr. Christopher Walsh discusses general principles of effective gene transfer for the hemophilias, specific information about viral vectors and non-viral gene transfer, and alternative target tissues for factor VIII and factor IX production. He highlights information about the immune response to gene transfer and reviews data from the hemophilia gene transfer trials to date. The future prospects for newer methods of therapy such as RNA repair and the use of gene-modified circulating endothelial progenitors are presented as possible alternatives to the more traditional gene therapy approaches. In Section II, Dr. Nigel Key focuses on inhibitor development in patients with hemophilia A. He reviews the progress in our understanding of the risk factors and presents newer information about the immunobiology of inhibitor development. He discusses the natural history of these inhibitors and the screening, laboratory diagnosis, and treatment, including the use of different modalities for the treatment of acute bleeding episodes. Dr. Key also presents information about the eradication of inhibitors by immune tolerance induction and reviews recent information from the international registries regarding the status and success of immune tolerance induction. In Section III, Dr. Margaret Rick discusses the diagnosis, classification, and management of von Willebrand disease. Attention is given to the difficulty of diagnosis in patients with mild bleeding histories and borderline laboratory test results for von Willebrand factor. She presents the value of different laboratory assays for both diagnosis and classification, and she relates the classification of von Willebrand disease to the choice of treatment and to the known genetic mutations. Practical issues of diagnosis and treatment, including clinical cases, will be presented.


2014 ◽  
Vol 22 (6) ◽  
pp. 1139-1150 ◽  
Author(s):  
Xiaomei Wang ◽  
Babak Moghimi ◽  
Irene Zolotukhin ◽  
Laurence M Morel ◽  
Ou Cao ◽  
...  

PLoS ONE ◽  
2009 ◽  
Vol 4 (8) ◽  
pp. e6376 ◽  
Author(s):  
Ashley T. Martino ◽  
Sushrusha Nayak ◽  
Brad E. Hoffman ◽  
Mario Cooper ◽  
Gongxian Liao ◽  
...  

2009 ◽  
Vol 9 (2) ◽  
pp. 104-114 ◽  
Author(s):  
Paul LoDuca ◽  
Brad Hoffman ◽  
Roland Herzog

Sign in / Sign up

Export Citation Format

Share Document