scholarly journals The Severe Spontaneous Bleeding Phenotype in a Novel Hemophilia a Rat Model with an Inversion Mutation Is Rescued By Platelet-Targeted FVIII Expression

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 219-219
Author(s):  
Qizhen Shi ◽  
Jeremy G Mattson ◽  
Scot A Fahs ◽  
Robert R Montgomery

Abstract Previous studies by our group and others have demonstrated that platelet targeted FVIII expression can rescue the bleeding phenotype in hemophilia A (HA) mice in both the inhibitor and non-inhibitor models. The clinical efficacy has been further proven in a dog HA non-inhibitor model. While these are informative, neither the canine nor mouse model entirely mimic human disease because 1) neither HA mice nor dogs exhibit the frequent spontaneous bleeding in severe HA patients, and 2) canine platelets do not contain FVIII carrier protein VWF and therefore store and release FVIII differently, thus precluding examining the efficacy of platelet gene therapy in the presence of inhibitors. The efficacy of platelet-FVIII gene therapy, therefore, needs to be examined in an animal model more representative of human disease. Since the inversion of FVIII gene is the most common mutation that causes severe HA in patients, using a CRISPR/Case9 strategy we generated a novel HA rat model in a Dahl/Salt Sensitive (Dahl) inbred congenic background (Dahl-F8KO) with the nearly entire murine FVIII gene inverted causing a translational stop 6 amino acids after the signal sequence of FVIII. There is no detectable FVIII in plasma in Dahl-F8KO rats (LOD is 1 U/dL). Spontaneous bleeding in the soft tissue, muscles, or joints occurred in 56.5% of homozygous affected animals (n=46) by the age of 8-weeks and up to 82.6% by the age of 32-weeks. This was significantly higher (P<0.0001) than the occurrence of spontaneous bleeding in another colony in Sprague Dawley (SD) background developed by Nielsen et al. using zinc finger nuclear strategy (J Thromb Haemost 2014) with a 13-bp deletion in exon 16 of FVIII. In SD-F8KO rat model, spontaneous bleeds occur in 23.3% and 42.1% of homozygous affected animals (n=85) that were housed in our facility by the age of 8- and 32-weeks, respectively. Likewise, the percentages of homozygous affected animals with 2, 3, or 4 bleeding episodes in the Dahl-F8KO colony were significantly higher than those in the SD-F8KO colony. In contrast, none of the heterozygous (F8+/-) animals ever have a spontaneous bleed. Similar to SD-F8KO rats, Dahl-F8KO rats can develop anti-FVIII inhibitory antibodies after 2 doses of recombinant human FVIII (rhF8) treatment. Thus, our novel Dahl-F8KO rat model is a unique animal model with the severest spontaneous bleeding phenotype yet reported. To investigate if platelet-derived FVIII can rescue the spontaneous bleeding phenotype in HA rats, we generated transgenic rats (2bF8Tg) in both SD- and Dahl-background in which FVIII expression is under control of the platelet-specific αIIb promoter (2bF8) using lentivirus-mediated oocyte transduction transgenesis. Bone marrow (BM) mononuclear cells were isolated from femurs of 2bF8Tg rats and transplanted into F8-/- rats preconditioned with 950 R TBI. Animals were closely monitored after BM transplantation (BMT), and blood samples were collected for FVIII assays and whole blood thrombin generation assay (wbTGA). After BMT from SD-2bF8Tg rats, no spontaneous bleeding was observed in SD-F8KO recipients within the entire study course (1 year at the time of this report) with a sustained platelet-FVIII expression of 20.9±8.1 mU/108 platelets (n=3). Similarly, we performed BMT from Dahl-2bF8Tg rats into Dahs-F8KO and found that 1 of 3 recipients had a bruise at the early stage of BM reconstitution. However, no any other spontaneous bleeding has yet been observed (5 months). To confirm that the bleeding diathesis in F8-/- rats was ameliorated after 2bF8 gene expression, wbTGA was performed. All parameters, including Lag Time, Peak Time, Peak Thrombin, Endogenous Thrombin Potential in 2bF8Tg-BMT recipients were significantly different compared to F8-/- control rats, but were similar to those obtained in WT control animals. Of note, neither detectable levels of plasma FVIII nor anti-F8 antibodies were detected in F8-/- recipients after receiving BMT from 2bF8Tg rats. In summary, we have developed a novel HA rat model with both the pathophysiology and clinical phenotype found in severe HA patients, which could be an ideal model for evaluating the clinical efficacy of new therapeutics. Our studies demonstrated that transplantation of BM cells that are genetically modified to express FVIII only in platelets can efficiently prevent the severe spontaneous bleeding in HA rats with no anti-F8 antibody development. Disclosures Montgomery: BCW: Patents & Royalties: GPIbM assay patent to the BloodCenter of Wisconsin.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5543-5543
Author(s):  
Steven C. Chen ◽  
Daniel Greenberg ◽  
Peiqing Ye ◽  
Earl W. Davie ◽  
Carol H. Miao

Abstract A major problem for clinical treatment of hemophilia A using factor replacement therapy is the high frequency formation of inhibitory antibodies against factor VIII. This problem is also predicted to occur following strategies currently aimed at targeted genetic correction of this disease. Recombinant activated factor VII (rFVIIa, NovoSeven®, Novo Nordisk, Bagsvaerd, Denmark) has been successfully used as an effective alternative treatment for hemophilia patients who have developed inhibitors. In order to decrease the cost and fluctuation of FVIIa levels associated with frequent infusions of rFVIIa, nonviral gene transfer of factor VII (FVII) was attempted in a hemophilia A mouse model. In addition, to investigate the potential thrombotic risks associated with prolonged, high level of FVIIa expression following gene transfer, we compared the effects of gene transfer vectors encoding zymogen FVII with an engineered secreted FVIIa in hemophilia A mice. We inserted murine FVII (mFVII) cDNA into a liver-specific vector developed recently in our lab to generate pBS-HCRHPI-mFVII-A. The mFVII cDNA sequence was then modified by site-directed mutagenesis to insert a protease cleavage site in between Arg152-Ile153. The resulting construct of murine FVIIa (mFVIIa) encodes a modified protein, which can be cleaved by intracellular proteases of the furin family to secrete the activated form of mFVII. Fifty mg of either mFVII or mFVIIa the plasmid was delivered into hemophilia A mouse liver by a rapid, high volume (hydrodynamics-based) infusion method (n=8 mice/group). Phenotypic correction was evaluated using tail-clip assays by measuring the hemoglobin levels in the collected blood. This assay indicated that vectors carrying either mFVII or mFVIIa can partially correct clotting following gene transfer. Significant correction in PT and APTT was observed in mFVII or mFVIIa-treated mice. Plasma-based and platelet-based thrombin generation (TG) assays indicated that either mFVII or mFVIIa significantly shorten peak time and peak thrombin levels of TG. In addition, these effects lasted long-term in plasmid treated mice for at least three months (experimental duration). We also constructed vectors carrying human FVII (hFVII) or FVIIa (hFVIIa) to test their efficacy for correcting hemophilia A. Constructs produced supra-physiological levels (800–1200ng/ml) of hFVII or hFVIIa assayed by ELISA at days 1 and 4 post-injection. However, the levels of hFVII and hFVIIa dropped precipitously at day 14 post-injection shown by both ELISA and Western blot, most likely due to the formation of species-specific antibodies against hFVII. These results clearly demonstrate that nonviral gene therapy of either zymogene FVII or activated FVII can rescue bleeding in hemophilia A mice.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1144-1144 ◽  
Author(s):  
Graça Almeida-Porada ◽  
Jyoti Desai ◽  
Charles Long ◽  
Mark Westhusin ◽  
Vladimir Pliska ◽  
...  

Abstract Hemophilia A, or Factor VIII (FVIII) deficiency, is the most common severe hereditary coagulation disorder, affecting 1 in 5000 male live births. Animal models in dog, mouse, and rabbit have been developed and used to study FVIII function and to evaluate new methods of treatment and prevention of inhibitor formation. Unfortunately, for unknown reasons, results obtained using these models didn’t always result in successful therapies when applied to humans. For new treatments to be safely and successfully translated from surrogate models to clinical trials, it is critical to develop an animal model that simultaneously and accurately parallels normal human physiology while mimicking human hemophilia’s physiopathological process. Due to its striking physiological and anatomical similarities to humans, sheep are considered an ideal model to study a vast array of pathologies. The aim of these studies was to re-establish, study, and characterize an extinct line of sheep with a spontaneous bleeding disorder that closely recapitulated human hemophilia A (ThrombHaemost68:618,1992). Thus, we used frozen semen from an affected male to generate hemophilia A carriers. We obtained 20 females that when compared to pooled control sheep plasma, exhibited slightly increased PTT levels (38.1±1.1; N=30s), normal PT and platelet number, and slightly decreased FVIII:C (70±3%). Levels of Fibrinogen, FIX, vWF activity and vWF:ag were also normal. A second round of reproductive manipulations using the carriers’ oocytes and the affected semen produced 23 more animals, 16 of which were obligate carriers with a similar phenotype. The other 8 animals exhibited prolonged bleeding from the umbilical cord that promptly stopped upon administration of purified human FVIII concentrate using recommended dosing. Due to the unfeasibility of clamping the umbilical cord, therapy with human FVIII was continued each 12 hours until the umbilical cord dropped off. Blood collected prior to the administration of FVIII showed that these animals had almost non-existent levels of FVIIIc, and an extremely prolonged PTT (91.5±2.9, N=30.3) with normal levels of platelets, fibrinogen, FVII, FIX, and vWF. 2 of the animals died shortly after birth due to extensive hematomas related to lambing trauma. The other 6 animals, now 5 months old (maturity 6–9 months), developed clinical symptomatology closely mimicking that of human patients with severe hemophilia A. Each of these animals had between 2–6 episodes of severe bleeding including hemarthroses of the elbow, shoulder, hip, and knee, multiple muscle hematomas, including 1 hematoma of the tongue and 1 episode of mild hematuria. All of the bleeding episodes resolved upon administration of 1–2 treatments with human FVIII. Animals have thus far received between 964-4546U of human FVIII. Of interest is that low-titer inhibitors (1.3; 2.6; 3 BU) were detected in 3 of the animals showing that the nature of the mutation present in these sheep renders them prone to inhibitor development. Characterization of the mutation is currently underway. We hope that this large animal model will contribute to a better understanding of hemophilia and the development of novel treatments that can directly translate to human patients, such as stem cell transplantation and gene therapy-based approaches.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1067-1067
Author(s):  
Juan Chen ◽  
Jocelyn Schroeder ◽  
Xiaofeng Luo ◽  
Robert R Montgomery ◽  
Qizhen Shi

Abstract Platelets play fundamental roles in hemostasis through surface proteins, e.g. GPIb, and storage proteins, such as von Willebrand factor (VWF). Our previous studies have demonstrated that FVIII ectopically targeted to platelets under control of the platelet-specific aIIb promoter (2bF8) is therapeutic in hemophilia A mice even in the presence of anti-FVIII inhibitory antibodies (inhibitors). Our recent studies have shown that VWF is essential in platelet-targeted FVIII gene therapy of hemophilia A with inhibitors. Without VWF, the clinical efficacy of platelet-FVIII was aborted. In primary hemostasis, platelets adhere to the vessel wall through the binding of GPIb to VWF. Thus, we wanted to explore whether GPIb is critical for maintaining the clinical efficacy of platelet-FVIII gene therapy of hemophilia A in the presence of inhibitors. To address this question, recipient FVIIInull (F8null) mice were immunized with recombinant FVIII (rhF8) to induce anti-FVIII inhibitory antibody development (the inhibitor model). Hematopoietic stem cells (HSCs) from GPIba knockout (Ibnull) mice were transduced with 2bF8 lentivirus and transplanted into rhF8-primed F8null mice under 1100 cGy total body irradiation (2bF8-Ibnull/F8null). As an inhibitor model control (2bF8-F8null/F8null), HSCs from F8null mice were transduced with 2bF8 and transplanted into rhF8-primed recipients under the same preconditioning. After bone marrow transplantation and reconstitution, animals were assessed for transgene expression and phenotypic correction. PCR analysis showed that the 2bF8 transgene was detected in all of the transduced recipients, demonstrating the viability of 2bF8-transduced engraftment. The level of functional platelet-FVIII expression as determined by a Chromogenic assay was 6.37 ± 5.62 mU/108 platelets (n = 6), which appeared to be higher than the level obtained in the inhibitor control group (1.97 ± 0.96 mU/108 platelets, n = 5), but there was no significant difference between these two groups. Since the size of Ibnull platelets is larger and the platelet number is lower than those in F8null mice, we converted FVIII levels into mU/ml whole blood and the level was 10.36 ± 5.33 mU/ml in 2bF8-Ibnull/F8null mice, which is not significantly different from the 2bF8-F8null/F8null mice (9.04 ± 3.38 mU/ml). When the tail bleeding test was used to grade phenotypic correction of the F8null coagulation defect, all transduced recipients in both groups survived beyond a 6-hour tail clipping test and the remaining hemoglobin level in the 2bF8-Ibnull/ F8null group was 46.4 ± 8.5%, which is not significantly different from the 2bF8-F8null/F8null group (48.3 ± 9.7%). The remaining hemoglobin levels in both groups were significantly higher than in the untransduced F8null control, in which only 5 of 12 animals survived beyond the 6-hour tail clipping test with a remaining hemoglobin level of 34.8 ± 7%. Thus, our data suggest that a lack of platelet GPIba does not negate the clinical efficacy of platelet FVIII gene therapy in hemophilia A in the presence of inhibitors. Disclosures Montgomery: Baxter: Consultancy; Bayer: Consultancy; CSL: Consultancy; Biogen: Consultancy; Octapharma: Consultancy; Grifols: Consultancy. Shi:BloodCenter of Wisconsin: Patents & Royalties: METHOD OF INDUCING IMMUNE TOLERANCE THROUGH TARGETTED GENE EXPRESSION..


Author(s):  
Mira Tafa ◽  
Sevim Naz Karışık ◽  
Ece Begüm Aksoy ◽  
Rüya Aslan

Cystic Fibrosis is a rare genetic disease that affects the transmission of chloride ions due to mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene. Even though there are nearly 2000 mutations identified to be related to the condition, the most common mutation is F508del; deletion of a phenylalanine residue at 508. On the other hand, G542X which is a Class I mutation is also found very commonly and there are no modulator treatments available for it. Furthermore, it was investigated that R553X mutation can as well be corrected simultaneously with G542X mutation. Therefore, the main focus is on designing a gene therapy project that can correct all these three mutations at once by utilizing the prime editing technique via lipid-based delivery. In this way, the mutations can be edited through plasmids that were designed containing 2 pegRNAs and the Cas enzyme. To implement such an approach efficiently, both ex vivo, an animal model, and in vivo steps are to be designed. For the cell line, fibroblasts are selected due to their simplicity and low cost. The animal model of the experiment is determined to be a ferret concerning the high similarity to the human's CFTR protein and finally, the procedure will follow on a direct application in human Cystic Fibrosis patients. The plasmids are thought to be delivered through a cationic liposome that will reach the lungs with the aid of a nebulizer. At the last stage of the experimental procedure, Sanger Sequencing will be done to see if the desired edit within the CFTR has been performed successfully, and Next Generation Sequencing will be executed to see if there has been an off-target mutation in the remainder of the genome. Whereas for detecting the presence and expression of CFTR protein in humans, immunodetection with flow cytometry will be conducted.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3286-3286 ◽  
Author(s):  
Qizhen Shi ◽  
Scot A. Fahs ◽  
David A. Wilcox ◽  
Hartmut Weiler ◽  
Sandra L. Haberichter ◽  
...  

Abstract Our laboratory has had a longstanding interest in the association of FVIII with its carrier protein, VWF, - both in plasma and within platelets and endothelial cells where there is co-storage with endogenous VWF when FVIII expression is induced. Recently we have demonstrated clinical efficacy of FVIII ectopically expressed and stored within platelets even though FVIII remains undetectable in plasma in the platelet-specific FVIII (2bF8) transgenic mice. This correction is maintained even in the presence of high titer inhibitors (JCI, 2006). Since VWF is also synthesized and stored in endothelial cells, FVIII could also be supplied together with VWF using cell-specific expression within a second vascular cell, providing local as well as systemic levels of FVIII. We developed another transgenic mouse in which FVIII was driven by the endothelial cell-specific Tie2 promoter (Tie2-F8) and contrasted it with the 2bF8 transgenic mouse previously described. When bred into the FVIIInull mouse, homozygous Tie2F8 mice maintained normal plasma FVIII levels (1.15 ± 0.16 U/ml) and 50% levels in Tie2F8+/− mice (0.56 ± 0.16 U/ml). Both Tie2F8+/− and Tie2F8+/+ phenotypes effectively abrogate the bleeding phenotype in FVIIInull mice. Since both cell types could be advantageous in the local delivery of FVIII at sites of injury, we explored the phenotypic correction of our transgenic models in the presence of FVIII inhibitory antibodies (Ab) that were generated by either transplantation of spleen cells from highly immunized FVIIInull mice or in transgenic mice immunized with recombinant FVIII with adjuvant. In marked contrast to the platelet 2bF8 mice, the Tie2F8 mice’ ability to survive a minor tail wound was significantly decreased (21/27 vs 3/10, P < 0.01) in the presence of FVIII inhibitory Ab. After one immunization, the inhibitor titers in Tie2F8 mice were much higher (850 – 3,000 BU/ml) than in 2bF8 mice (10 – 150 BU/ml). Furthermore, in the presence of Ab, circulating FVIII levels in whole blood were maintained in the platelets of 2bF8 mice, but dropped to undetectable levels in the Tie2F8 mice. While both platelets and endothelial cells could provide local FVIII at sites of injury, in the presence of inhibitory Ab, endothelial delivery was not clinically efficacious and appeared to rely on the plasma FVIII levels it could maintain for its clinical efficacy. Thus, local delivery of FVIII by platelets concentrates its effect at sites of vascular injury, and storage within platelets protects FVIII from inactivation by Abs. This effect is a unique property of platelet delivery and is not maintained with endothelial FVIII delivery. Our further studies demonstrated when the lentivirus-mediated gene transfer system was used to introduce FVIII expression in either platelets or endothelial cells by transplantation of bone marrow cells transduced with 2bF8 lentivirus or systemic transduction of endothelial cells with Tie2F8 lentivirus, the outcome was different. Sustained phenotypic correction without Ab development was achieved in lentivirus-mediated platelet-specific gene therapy. In contrast, all endothelial-specific Tie2F8 lentivirus treated mice developed inhibitory Ab (50 ± 14 BU/ml) with no detectable plasma FVIII. Thus, we have demonstrated that platelets are potentially the optimal target for gene therapy of hemophilia A with inhibitors.


2014 ◽  
Vol 113 (11) ◽  
pp. 4005-4013 ◽  
Author(s):  
Ying Feng ◽  
Xin Zeng ◽  
Wei-hua Li ◽  
Wen-cong Wang ◽  
Li-si Ou-Yang ◽  
...  

Hematology ◽  
2019 ◽  
Vol 2019 (1) ◽  
pp. 1-8 ◽  
Author(s):  
Amit C. Nathwani

Abstract Gene therapy offers the potential for a cure for patients with hemophilia by establishing continuous endogenous expression of factor VIII or factor IX (FIX) following transfer of a functional gene to replace the hemophilic patient’s own defective gene. The hemophilias are ideally suited for gene therapy because a small increment in blood factor levels (≥5% of normal) is associated with significant amelioration of bleeding phenotype in severely affected patients. In 2011, the St. Jude/UCL phase 1/2 trial was the first to provide clear evidence of a stable dose-dependent increase in FIX levels in patients with severe hemophilia B following a single administration of adeno-associated viral (AAV) vectors. Transgenic FIX expression has remained stable at ∼5% of normal in the high-dose cohort over a 7-year follow-up period, resulting in a substantial reduction in spontaneous bleeding and FIX protein usage without toxicity. This study has been followed by unparalleled advances in gene therapy for hemophilia A and B, leading to clotting factor activity approaching normal or near-normal levels associated with a “zero bleed rates” in previously severely affected patients following a single administration of AAV vectors. Thus, AAV gene therapies are likely to alter the treatment paradigm for hemophilia A and B. This review explores recent progress and the remaining limitations that need to be overcome for wider availability of this novel treatment of inherited bleeding disorders.


2020 ◽  
Vol 4 (1) ◽  
pp. 55-65 ◽  
Author(s):  
Qizhen Shi ◽  
Jeremy G. Mattson ◽  
Scot A. Fahs ◽  
Aron M. Geurts ◽  
Hartmut Weiler ◽  
...  

Key Points A novel HA rat model caused by an inversion exhibits a severe spontaneous bleeding phenotype. The severe spontaneous bleeding phenotype in HA rats is rescued by platelet-targeted FVIII expression.


2018 ◽  
Vol 1 (5) ◽  
Author(s):  
Hainiu Wang

Objective Diabetes mellitus coexisting with hypertension can accelerate and increase the occurrence and development of cardiovascular disease, stroke, diabetic nephropathy and retinopathy, and the establishment of corresponding animal models can provide the clinical evidence for hypertension with diabetes mellitus. The model of diabetic rats with hypertension was established in order to find the most reasonable model. Methods  Using the method of literature, the key words in Pubmed were: "diabetes mellitus"; "hypertension" ; "rat" ; The qualifying language for animal model is English, with a limitation period of 2008-2018. A total of 157 papers were collected and included into the standard: ①exclude the relevant model of renal hypertension; ②No other diseases other than diabetes and hypertension; A total of 42 studies were included. Results  There are 6 models of diabetes mellitus with hypertension in common use at present. ①Surgically induced bilateral renal artery stenosis in rats, followed by low-dose STZ intraperitoneal injection and feeding high-calorie diet. ②special chemical (STZ or alloxan) directly injected into Spontaneously Hypertensive Rat (SHR) or combined with specific dietary induction; ③Hybridization of SHR and spontaneously diabetic rats; ④Inbred purebreds with a propensity to spontaneously diabetes, obese SHR and obese Zucker rats; ⑤Diabetes was induced by STZ injection with SHR in primary hypertensive rats. ⑥High-fat diet plus intraperitoneal injection of STZ into diabetic model combined with 1% NaCl water feeding. Conclusions The preparation methods of several models have their own advantages and disadvantages. From the point of view of the pathogenesis of human diseases, it is more ideal to make inbred model with the tendency of spontaneous diabetes. From the point of view of economy and economy, the low-dose STZ intraperitoneal injection of SHR into diabetic hypertensive rat model is low cost, high model rate and convenient. The model rats with type 2 diabetes mellitus and hypertension were induced by high-fat diet plus STZ plus 1% NaCl by intraperitoneal injection of STZ in combination with drinking water. Relatively close to the course of human disease, the cost is lower, and it is the most widely used modeling method at present. Conclusion: Different research targets correspond to different animal model vectors. In the future, we should try to establish a more perfect model of diabetes mellitus with hypertension, which is more similar to human disease, so as to provide a good platform for disease prevention and treatment.


Sign in / Sign up

Export Citation Format

Share Document