{BLR 2843} Abgenix-American Biogenetic Systems-Aquasearch-Beaufour Ipsen-Bioprospecting-Cancer-CDC-Cell Genesys-Chlamydia-Cryptosporidium-Dengue Fever-Diversa-Eos-Factor VHI-GalaGen-Gemini-Gene Therapy-Genset-Genzyme-GVAX-Hemophilia A-Herpesvirus 8-IGEN-Immune Tolerance-Immunotech-Japan Tobacco-Marine Organisms-Medicines Company-Mexico-Migraines-Neoprobe-Neuroblastoma-Nutritional Products-Obesity-Octagen-OraVax-p53-Pasteur Merieux Connaught-PMCI-Prostate Cancer-Radiotracers-Rapigene-Research Corp. Technologies-Schering-Plough-SNPs-University of Maryland-Vaccines-Wyeth-Ayerst

1999 ◽  
Vol 18 (1) ◽  
pp. 61-63
Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4170-4170
Author(s):  
Yingyu Chen ◽  
Erin L. Kuether ◽  
Jocelyn A. Schroeder ◽  
Robert R. Montgomery ◽  
David W. Scott ◽  
...  

Abstract Abstract 4170 Our previous studies have shown that targeting FVIII expression to platelets (2bF8) can correct murine hemophilia A phenotype even in the presence of inhibitory antibodies. In the present study, we wanted to explore 1) whether platelets containing FVIII can act as an immunogen; and 2) whether platelet-derived FVIII can induce immune tolerance in a hemophilia A mouse model. To investigate whether platelets containing FVIII can act as an immunogen in hemophilia A mice, we infused transgenic mouse platelets with a level of platelet-FVIII of 6 mU/108 platelets to naïve FVIIInull mice weekly for 8 weeks. These platelets were between 30 to 50% of total platelets upon infusion and the levels of platelet-FVIII in the infused animals were 0.11 ± 0.01 mU/108 platelets (n = 6) one week after infusion. No anti-FVIII inhibitory antibodies were detected in the infused mice during the study course. All animals developed inhibitors following further challenged with recombinant human FVIII (rhFVIII) at a dose of 50 U/kg by intravenous injection weekly for 4 weeks, indicating that infusion of platelets containing FVIII does not trigger immune response in hemophilia A mice. To explore whether platelet-derived FVIII will act as an immunogen in the presence of primed spleen cells (from mice already producing inhibitory antibody), we co-transplanted splenocytes from highly immunized FVIIInull mice and bone marrow (BM) cells from 2bF8 transgenic mice into 400 cGy sub-lethal irradiated FVIIInull recipients. We monitored the levels of inhibitory antibodies in recipients for up to 8 weeks and found that inhibitor titers declined with time after transplantation. We then challenged co-transplantation recipients with rhFVIII and found that inhibitor titers in the control group co-transplantat of FVIIInull BM cells increased 103.55 ± 64.83 fold (n = 4), which was significantly more than the group receiving 2bF8 transgenic BM cells (14.34 ± 18.48, n = 5) (P <.05). The inhibitor titers decreased to undetectable in 40% of 2bF8 transgenic BM cells co-transplantation recipients even after rhFVIII challenge, indicating immune tolerance was induced in these recipients. To further explore the immune response in the lentivirus-mediated platelet-derived FVIII gene therapy of hemophilia A mice, we transduced hematopoietic stem cells from pre-immunized FVIIInull mice with 2bF8 lentivirus (LV) followed by syngeneic transplantation into pre-immunized lethally irradiated FVIIInull recipients and monitored the levels of inhibitor titers in recipients. After full BM reconstitution, platelet-FVIII expression was sustained (1.56 ± 0.56 mU/108platelets, n = 10), but inhibitor titers declined with time, indicating that platelet-derived FVIII does not provoke a memory response in FVIIInullmice that had previously mounted an immune response to rhFVIII. The t1/2 of inhibitor disappearance in 2bF8 LV-transduced recipients (33.65 ± 11.12 days, n = 10) was significantly shorter than in untransduced controls (66.43 ± 22.24 days, n = 4) (P <.01). We also transplanted 2bF8 LV-transduced pre-immunized HSCs into 660 cGy sub-lethal irradiated naïve FVIIInull mice. After BM reconstituted, recipients were assessed by platelet lysate FVIII:C assay and tail clip survival test to confirm the success of genetic therapy. Animals were then challenged with rhFVIII. Only 2 of 7 2bF8 LV-transduced recipients developed inhibitory antibodies (55 and 87 BU/ml), while all untransduced control developed high titer of inhibitors (735.50 ± 94.65 BU/ml, n = 4). In conclusion, our results demonstrate that 1) platelets containing FVIII are not immunogenic in hemophilia A mice; and 2) platelet-derived FVIII may induce immune tolerance in hemophilia A mice with or without pre-existing inhibitory antibodies. This tolerance induction would add an additional significant benefit to patients with platelet-derived FVIII gene therapy strategy because protein infusion could be administered in some special situations (e.g. surgery in which a greater levels of FVIII may be required) with minimized risk of inhibitor development. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 243-243 ◽  
Author(s):  
Jonathan D Finn ◽  
Denise E Sabatino ◽  
Margareth C Ozelo ◽  
Shangzhen Zhou ◽  
David Lillicrap ◽  
...  

Abstract The formation of antibodies (or inhibitors) to FVIII is a major complication in the treatment of humans with hemophilia A (HA), affecting up to 30% of individuals with severe or moderate disease. Inhibitors develop mainly in young boys, and render the treatment with infused protein suboptimal or completely ineffective. Inhibitor patients exhibit significant delays in their maturation and physical development. Thus, inhibitor formation negatively affects both hematological and systemic outcomes in these patients and, therefore the prevention or eradication of inhibitors is of fundamental importance. To eliminate the presence of inhibitors in hemophilia, an immune tolerance induction (ITI) is followed consisting of large amounts of FVIII injected on a daily basis for long periods of time (months to years) with variable success rates. Although liver-directed gene therapy has been used to deliver therapeutic transgenes and can induce tolerance to the expressed protein, to date there have been no large animal studies using liver gene therapy to eradicate inhibitors to FVIII. The HA dog colonies are an excellent model for studying tolerance induction as both colonies have the same mutation found in the majority of the human population (inversion of intron 22) resulting in severe hemophilia. We hypothesize that sustained expression of cFVIII could mimic the effects of ITI in eradicating inhibitors to canine FVIII in dogs that have a history of inhibitors, thus demonstrating a potential alternative to the current ITI protocol. We injected AAV-8 vectors encoding the light and heavy chain of canine FVIII (cFVIII) (two separate vectors) driven by a liver specific promoter (2.5 e13 vg/kg per vector) into the peripheral vein of four adult (7–36 months old) HA dogs with inhibitors. The first dog treated (K03, 12 months old) was from a new strain from the Chapel Hill (UNC) HA dog colony prone to inhibitor formation. K03 had a historical maximum inhibitor titer of 12 B.U. and a titer of 3 B.U. at the time of vector infusion. Post AAV injection, we documented a slow shortening of whole blood clotting time (WBCT) to normal values (13 min) and inhibitors decreased to undetectable levels by week 3. K03 has demonstrated cFVIII antigen levels (~30 ng/mL) that are stable out to 250 days post treatment. When challenged with 4 weekly injections of 125 μg of highly purified recombinant cFVIII he failed to generate an inhibitor, indicating induction of immune tolerance. K01 (UNC, 20 months old) had a historical maximum inhibitor titer of 12–13 B.U. and a titer of 3 B.U. at the time of vector infusion. We observed an initial transient rise in cFVIII antigen, followed by a decrease to baseline that corresponded to an increase in inhibitor titer to 5–6 B.U. before decreasing to undetectable levels within 4 weeks of gene transfer. cFVIII antigen levels have slowly increased over time, and have reached 14 ng/mL by day 150. WBCT has decreased corresponding with decreasing inhibitor titers and has normalized to 13.5 min. L44 (UNC, 8 months old) had a historical maximum inhibitor titer of 3 B.U. and a titer of 1.5 at the time of infusion. After vector infusion we observed a rapid onset of cFVIII expression (peaking at 50 ng/mL) that decreased to near baseline by day 8 before increasing and stabilizing at 15–20 ng/ml (out to 40 days) and inhibitor titers have decreased while WBCT has decreased to 18 min. The fourth dog (Wembley, 3 years old) is from the HA dog colony at Queens University and had previously been exposed to both canine and human FVIII and presented inhibitors to both human (10 B.U.) and canine (3.6 B.U.) FVIII. After vector infusion Wembley showed a transient increase in cFVIII levels, followed by a decrease in cFVIII expression and no indication of tolerance induction after 10 weeks, ongoing observation. Notably, this dog presented high titer IgG1 and IgG2 anti-cFVIII antibodies, while the other three dogs had mostly IgG2 (equivalent to human IgG4). No abnormalities in liver or renal functions were observed in these dogs. Factors such as exposure to xeno-antigens, the nature of the antibody response, duration of inhibitor or age may influence the outcome of the tolerance induction protocol. Collectively, these data demonstrate the potential of liver directed, AAV mediated gene delivery to no only treat genetic deficiencies such as hemophilia, but to induce immune tolerance to the transgene in the setting of pre-existing inhibitory antibodies.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 13-13
Author(s):  
Debalina Sarkar ◽  
David M Markusic ◽  
Cox Terhorst ◽  
Todd Brusko ◽  
Roland W Herzog

Abstract Abstract 13 Hemophilia A and B result from deficiency in clotting factor VIII (FVIII) or IX (FIX), respectively. In a subset of patients, treatment by factor replacement therapy is limited by formation of inhibitory antibodies to the clotting factors, representing a serious complication that increases risks of morbidity and mortality. Immune responses to the therapeutic coagulation factors are also a concern in newly emerging gene therapies. Regulatory T cells (Treg) offer to be a novel alternative pathway toward immune tolerance. Treg has been identified as a major component of immune tolerance to coagulation factors in pre-clinical studies. Therefore, we hypothesize that ex vivo expanded autologous Treg can suppress inhibitor formation. Our study seeks to test this approach in hemophilic mice. Initially, we optimized in vitro expansion of murine BALB/c-derived Treg. Using flow sorting, GFP+ cells were purified (>98% purity) from spleens of BALB/c knock-in mice containing a GFP reporter linked to FoxP3 expression with an IRES sequence. Sorted cells were stimulated in culture using anti-CD28/anti-CD3 beads in the presence of high-level IL-2 (1000 U/ml). IL-2 was replenished every second day in culture. After ∼1 week, cells were freshly stimulated. At the end of 2 weeks, viability, purity, and FoxP3/GFP expression was confirmed. Greater than 30-fold expansion was repeatedly accomplished. Assumming a dose of 1×106 Treg/mouse, expansion is sufficiently robust to treat >30 mice starting with Treg from 2–3 donor mice. Ex vivo expanded Tregs were adoptively transferred to male hemophilia A mice (BALB/c F8e16 −/−), which were then treated with F.VIII (1 IU human B domain-deleted F.VIII, IV, once per week) for two months. Bethesda assays demonstrated that Treg transplant had effectively suppressed inhibitor formation. Inhibitor titers in control mice were 15–20 BU at 1 month and 30–40 BU at two months. In contrast, Treg treated mice (n=5 per group) formed at most low-titer inhibitors (2–3 BU for both time points). By 2 months, peripheral Treg frequencies had returned to near baseline. To further demonstrate presence of a Treg population capable of suppressing antibody formation against F.VIII, a secondary transfer of sorted CD4+CD25+ splenocytes was performed. Recipient hemophilia A mice were immunized against F.VIII in adjuvant. Compared to mice receiving control Treg, there was significant (P<0.05) suppression of inhibitor formation against F.VIII. In other experiments, Treg therapy was also able to significantly reduce inhibitor titers in hemophilia A mice with pre-existing F.VIII inhibitors. We chose hemophilia A mice (n=6) that developed on average 25 BU from F.VIII replacement therapy. Half of these received Treg transplant, and all mice received 8 more weeks of F.VIII treatment. Inhibitor titers in the control group increased to ∼100 BU. Treg therapy substantially reduced this response (to 15–20 BU, P<0.001). In order to evaluate Treg therapy for hemophilia B, BALB/c F9 −/− × FoxP3-GFP mice were generated. Treg were isolated from these mice as described above, expanded in vitro, and transferred to (BALB/c F9 −/−) hemophilia B mice. Two days later, to test their effectiveness in a gene therapy setting, these mice were treated by intramuscular injection of AAV1 vector expressing human F.IX. By 6 weeks after gene transfer, control mice had formed high-titer antibody against hF.IX (>20 mg IgG/ml plasma, ∼ 10 BU). In contrast, anti-hF.IX formation was undetectable in mice that had received Treg prior to vector administration (n=4/group). While perhaps not as potent as antigen-specific Treg, our data demonstrate the ability of highly purified and ex vivo expanded bulk Treg to control inhibitor formation and thus support their utility for tolerance induction in hemophilia. Their effectiveness may involve emergence of a more specific Treg population after repeated in vivo exposure to antigen. In gene therapy, Treg transplant may be a more desirable alternative to use of immune suppressive drugs. Providing additional immune regulation around the time of vector administration, i.e. when activation signals are provided to the immune system, could be sufficient to prevent immune rejection long-term while inducing antigen-experienced Treg for durable tolerance. Disclosures: Herzog: Genzyme Corp.: Royalties, AAV-FIX technology, Royalties, AAV-FIX technology Patents & Royalties.


2006 ◽  
Vol 13 ◽  
pp. S419
Author(s):  
Hideto Matsui ◽  
Masaru Shibata ◽  
Brian Brown ◽  
Andrea Labelle ◽  
Carol Hegadorn ◽  
...  

2017 ◽  
Vol 15 (10) ◽  
pp. 1994-2004 ◽  
Author(s):  
Y. Chen ◽  
X. Luo ◽  
J. A. Schroeder ◽  
J. Chen ◽  
C. K. Baumgartner ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document