Gene Transfer of Canine FVIIa Results in Partial Correction of Canine Hemophilia: A Model for FVIII/FIX Gene-Based Bypassing Agents.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 195-195 ◽  
Author(s):  
Paris Margaritis ◽  
Elise Roy ◽  
Harre D. Downey ◽  
Shangzen Zhou ◽  
Elizabeth Merricks ◽  
...  

Abstract Despite its extensive use particularly in the management of hemophilic inhibitor patients, recombinant Factor VIIa (rhFVIIa) infusion has important limitations stemming from the nature of FVIIa itself, since its short half-life necessitates repeated injections and also carries high treatment costs. To overcome these, we have designed a gene transfer approach using a modified FVII transgene that is cleaved intracellularly and secreted in the active form, FVIIa. Using the human and murine analogue of this engineered transgene we have shown phenotypic correction of hemophilia B mice, following adeno-associated virus (AAV) - mediated, liver-directed gene delivery (Margaritis et al., 2004). In order to demonstrate efficacy in a large animal model of hemophilia, we cloned the canine Factor VII cDNA and generated the canine homologue of our modified transgene (cFVIIa). Recombinant cFVII zymogen and cFVIIa were purified and characterized in vitro in a clotting-based assay using canine reagents only (activated partial thromboplastin time [aPTT]). We found that cFVIIa had activity indistinguishable from rhFVIIa, while cFVII zymogen had negligible activity (5% rhFVIIa). In order to demonstrate in vivo efficacy, we produced 4 lots of an AAV8-based vector directing liver-specific expression of cFVIIa with similar vector titers (2–5 E13 vector genomes [vg]/ml). In hemophilia A (HA) or B (HB) mice, tail-vein delivery of 0.3 – 1.2 E12 vg/mouse (1.2 – 4.8 E13 vg/kg) resulted in long-term normalization of the hemophilic phenotype, demonstrating that cFVIIa can correct the defect in murine hemophilia. We proceeded to infuse 4 hemophilia dogs, with increasing vector doses: HB male (2.06 E13 vg/kg); HA male (6.25 E13 vg/kg); HA female (1.25 E14 vg/kg); HA male (1.25 E14 vg/kg). None of the dogs showed any adverse effects following vector delivery at any dose (the initial HB dog has been followed for almost 2 years [ongoing]). We followed the level of gene expression by clotting assays (prothrombin time [PT]/aPTT) and whole blood clotting time (WBCT). The initial dose of 2.06 E13 vg/kg resulted in a transient reduction in the PT/aPTT/WBCT. A considerable and sustained reduction in PT (18 sec, normal is ∼25 sec), aPTT (19 sec, normal is ∼30 sec, hemophilic is >40sec) and WBCT (25min, normal is ∼15min, hemophilic is >40min) was observed following administration of 6.25 E13 vg/kg in an HA male dog. Two more HA dogs were infused with 1.25 E14 vg/kg (male and female). The female HA dog exhibited only a modest decrease in aPTT (22sec), despite the vector dose increase, and a reduction in WBCT (30min), an observation that could be due to previously described gender-specific effects on gene expression. From preliminary and ongoing observations, the male HA dog infused also exhibited a decrease in WBCT. As an efficacy endpoint, the dogs exhibited a total of 3 bleeding episodes (none likely to be spontaneous, occurred in the lowest dose HB dog) in a cumulative time period of 38.5 months, compared to the expected 16 episodes (Brunetti-Pierri et al., 2005). In summary, our results demonstrate for the first time that gene transfer using a Factor VIII/Factor IX bypassing agent (canine FVIIa) can result in partial correction of the hemophilic phenotype in a large animal model of hemophilia.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. lba-4-lba-4
Author(s):  
Paris Margaritis ◽  
Elise Roy ◽  
Majed N Aljamali ◽  
Harre Downey ◽  
Shangzhen Zhou ◽  
...  

Abstract Recombinant human activated Factor VII (rhFVIIa) is extensively used in the management of hemophilic inhibitor patients. However, the short plasma half-life and need for repeated infusions are amongst the limiting factors for its use in prophylaxis. In an attempt to overcome these, we have designed a gene transfer approach using continuous expression of a modified FVII transgene that is secreted in the active form, FVIIa. In proof-of-concept experiments, we previously demonstrated that Adeno-associated virus (AAV)–mediated gene transfer of the modified Factor VII gene can result in long-term phenotypic correction in hemophilia B mice. However, a prerequisite to a human application is the demonstration of efficacy as well as safety in a large animal model of hemophilia. Hence, we utilized the canine hemophilia model that closely resembles the human disease and has been shown to be an excellent predictor of efficacy of treatments for hemophilia in humans. As we previously reported (Blood 110 (11), p65a. 2007), we generated a canine FVII transgene that is expressed and secreted in a two-chain, activated form (cFVIIa). An AAV serotype 8 vector directing expression of this transgene was delivered via the portal vein into 4 hemophilia dogs, at vector doses of 2.06 E13 (one hemophilia B dog) − 1.25 E14 (three hemophilia A dogs) vector genomes (vg)/kg. The current abstract presents a rigorous characterization of the clotting parameters in the treated dogs, clinical safety and efficacy results of a substantially longer follow-up, and a laboratory safety profile of continuous expression of canine FVIIa. Gene expression in the treated dogs was followed by antigen and clotting assays (prothrombin time [PT], whole blood clotting time [WBCT]) as well as rotational thromboelastography (TEG). The initial dose of 2.06 E13 vg/kg resulted in a modest and transient reduction in the PT, WBCT and TEG parameters over a period of >31 months, accompanied by marginal changes in cFVIIa antigen levels. However, a considerable and sustained reduction in PT (supra-physiologic), WBCT (partial correction) and TEG parameters (near-normalization) was observed following administration of 6.25 E13 – 1.25 E14 vg/kg in three hemophilia A dogs in a cumulative total of 42 months of observation. Antigen levels in the treated hemophilia A dogs were in the range of 1–2.5 μg/ml. As untreated hemophilic dogs exhibit 5–6 spontaneous bleeds per year, we utilized this as an important efficacy endpoint. None of the treated dogs (including the lowest dose hemophilia B dog) exhibited any spontaneous bleeding episodes (34 expected within the observation period) in contrast to concurrently observed controls, which exhibited clinically detectable bleeding episodes at the predicted rate. Given the procoagulant nature of cFVIIa, the safety profile of this approach was thoroughly investigated. We did not observe any abnormal changes in serum chemistries, platelet counts, levels of D-dimer, fibrinogen or thrombin-antithrombin, arguing against a procoagulant state following cFVIIa-gene transfer. In summary, our results demonstrate for the first time that AAV8 vector doses of up to 1.25 E14 vg/kg (2.6 E15 vg total) are well-tolerated in hemophilic dogs; continuous expression of FVIIa at levels of 1–2.5 μg/ml is safe in hemophilic dogs, and can result in a sustained, measurable improvement in the clinical bleeding course of these animals. These data suggest that this model approach of a gene-based Factor VIII/Factor IX bypassing agent has therapeutic potential for hemophilia as has been shown for bolus infusion of rhFVIIa


Blood ◽  
2009 ◽  
Vol 113 (16) ◽  
pp. 3682-3689 ◽  
Author(s):  
Paris Margaritis ◽  
Elise Roy ◽  
Majed N. Aljamali ◽  
Harre D. Downey ◽  
Urs Giger ◽  
...  

Abstract Continuous expression of activated factor VII (FVIIa) via gene transfer is a potential therapeutic approach for hemophilia patients with or without inhibitory antibodies to human factor VIII (FVIII) or IX (FIX). Here, we investigate whether gene transfer of an engineered canine FVIIa (cFVIIa) transgene can affect hemostasis in a canine model of hemophilia, a good predictor of efficacy of hemophilia treatments. Purified recombinant cFVIIa exhibited 12-fold higher tissue factor–dependent activity than purified recombinant zymogen cFVII. Subsequently, we generated a serotype 8 recombinant adeno-associated viral vector expressing cFVIIa from a liver-specific promoter. Vector delivery via the portal vein in hemophilia A and B dogs was well tolerated, and long-term expression of cFVIIa resulted in a shortening of the prothrombin time, partial correction of the whole blood clotting time and thromboelastography parameters, and a complete absence of spontaneous bleeding episodes. No evidence of hepatotoxicity, thrombotic complications, or inhibitory immune response was found. These data provide the first evidence for in vivo efficacy and safety of continuously expressed FVIIa as a FVIII/FIX-bypassing agent in a large animal model of hemophilia, avoiding the risk of inhibitor formation associated with bolus FVIII or FIX infusion.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3525-3525 ◽  
Author(s):  
David A. Wilcox ◽  
Lily M. Du ◽  
Sandra L. Haberichter ◽  
Paula M. Jacobi ◽  
Juan Fang ◽  
...  

Abstract In order to develop a large animal model for platelet-targeted gene therapy of Hemophilia A, we transduced normal canine bone marrow with a lentivirus vector under the transcriptional control of integrin αIIb gene promoter driving expression of human BDD-FVIII. Bone marrow was collected from normal dogs and transduced in the presence of recombinant canine c-SCF, c-IL-6, human h-TPO, and h-flt-3/flk-2 ligand differentiation factor. Immunofluorescence analysis using antibodies against human FVIII, and the megakaryocyte-specific marker integrin αIIb revealed synthesis of FVIII within tissue cultured megakaryocytes derived from lentivirus transduced bone marrow cells. This result appeared similar to synthesis, trafficking and storage of FVIII trafficked to α-granules of the murine “small animal” model for hemophilia A and human megakaryoyctes in vitro. To examine the effect of FVIII expression in platelets, in vivo, FVIII-transduced canine bone marrow was xenotransplanted into immune-compromised “NOD-SCID” mice treated with a sublethal dose (350 cGy) of irradiation. Flow cytometric analysis using antibodies specific for canine glycoproteins Ibα and integrin αIibβ3 revealed that circulating canine platelets comprised approximately 20–30% of the total platelet population in whole blood isolated from the mice within four weeks after xenotransplant. Immunofluorescence confocal microscopy detected a punctuate staining for FVIII that co-localized with a platelet α–granule protein, P-selectin, within a subpopulation of canine platelets isolated from murine whole blood. In contrast, FVIII was not detected in platelets from control samples. In addition, chromogenic analysis of platelets isolated from mice transplanted with FVIII-transduced bone marrow demonstrated the presence of a biologically active form of FVIII, FVIII:C at 2 mU/ml/lysate of 1×108 platelets. These results indicate that canine megakaryocytes are able to synthesize and store a biologically active form of FVIII that can be retained within progeny platelets. We speculate that dogs affected with hemophilia A should serve as an ideal “large animal” model to test if FVIII can undergo regulated release from platelets following physiologic hemostatic response to vessel injury. This raises the possibility of developing a locally inducible secretory pool of FVIII in platelets of patients with Hemophilia A following autologous transplantation of FVIII-transduced hematopoietic stem cells.


2013 ◽  
Vol 5 (211) ◽  
pp. 211ra159-211ra159 ◽  
Author(s):  
L. Tilemann ◽  
A. Lee ◽  
K. Ishikawa ◽  
J. Aguero ◽  
K. Rapti ◽  
...  

2017 ◽  
Vol 1 (26) ◽  
pp. 2591-2599 ◽  
Author(s):  
Lindsey A. George

Abstract Concurrent with the development of recombinant factor replacement products, the characterization of the F9 and F8 genes over 3 decades ago allowed for the development of recombinant factor products and made the hemophilias a target disease for gene transfer. The progress of hemophilia gene therapy has been announced in 3 American Society of Hematology scientific plenary sessions, including the first “cure” in a large animal model of hemophilia B in 1998, first in human sustained vector-derived factor IX activity in 2011, and our clinical trial results reporting sustained vector-derived factor IX activity well into the mild or normal range in 2016. This progression to clinically meaningful success combined with numerous ongoing recombinant adeno-associated virus (rAAV)–mediated hemophilia gene transfer clinical trials suggest that the goal of gene therapy to alter the paradigm of hemophilia care may soon be realized. Although several novel therapeutics have recently emerged for hemophilia, gene therapy is unique in its potential for a one-time disease-altering, or even curative, treatment. This review will focus on the prior progress and current clinical trial investigation of rAAV-mediated gene transfer for hemophilia A and B.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5193-5193 ◽  
Author(s):  
Kristy L. Richards ◽  
Alison Motsinger-Reif ◽  
Hsiao-Wei Chen ◽  
Yuri D. Fedoriw ◽  
Cheng Fan ◽  
...  

Abstract Abstract 5193 Diffuse large B-cell lymphoma (DLBCL) affects ∼25,000 people in the U.S. each year, and fewer than half of them are cured with standard therapy. DLBCL can be divided into two subtypes by gene expression profiling, germinal center B-cell (GCB) type and activated B-cell (ABC) type. ABC-type DLBCL patients have significantly poorer outcomes. To improve therapeutic options, better animal models that accurately mimic human DLBCL (hDLBCL) are needed. Canine DLBCL is one of the most common cancers in veterinary oncology. Similar to human DLBCL patients, dogs with lymphoma are treated with both CHOP-like regimens and autologous stem cell transplants. Morphologically, canine lymphomas are similar to hDLBCL, with shared histologic markers, such as CD20 and PAX5. With recent technologies based on knowledge of the canine genome sequence, it is now possible to evaluate dogs as a potential large-animal model for hDLBCL. We evaluated 58 canine B-cell lymphomas by generating comprehensive gene expression profiles and comparing them to previously published hDLBCL expression profiles. Canine B-cell lymphoma expression profiles were similar in some ways to hDLBCLs. For instance, increased expression of NF-kB pathway genes was noted in a subset of lymphomas, mirroring NF-kB pathway activation in human ABC-type DLBCL. Furthermore, immunoglobulin heavy chain (IGH) mutation status, which is correlated with ABC/GCB cell of origin in hDLBCL, separated canine DLBCL into two groups with statistically different progression-free and overall survival times. However, canine DLBCL differed from hDLBCL in other aspects, including rare immunohistochemical positivity for BCL6 and MUM1/IRF4. Collectively, these results define aspects of canine B-cell lymphomas that resemble hDLBCL, identifying molecular similarities that could allow dogs to be used as a representative model of hDLBCL. Further comparative studies, including therapeutic trials, could potentially improve outcomes in both species. Disclosures: No relevant conflicts of interest to declare.


2011 ◽  
Vol 22 (7) ◽  
pp. 843-852 ◽  
Author(s):  
Patricia Favaro ◽  
Jonathan D. Finn ◽  
Joshua I. Siner ◽  
J. Fraser Wright ◽  
Katherine A. High ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document