Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
1.
Blood ; 137(6): 763-774, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33067633

ABSTRACT

Gene therapy has the potential to maintain therapeutic blood clotting factor IX (FIX) levels in patients with hemophilia B by delivering a functional human F9 gene into liver cells. This phase 1/2, open-label dose-escalation study investigated BAX 335 (AskBio009, AAV8.sc-TTR-FIXR338Lopt), an adeno-associated virus serotype 8 (AAV8)-based FIX Padua gene therapy, in patients with hemophilia B. This report focuses on 12-month interim analyses of safety, pharmacokinetic variables, effects on FIX activity, and immune responses for dosed participants. Eight adult male participants (aged 20-69 years; range FIX activity, 0.5% to 2.0%) received 1 of 3 BAX 335 IV doses: 2.0 × 1011; 1.0 × 1012; or 3.0 × 1012 vector genomes/kg. Three (37.5%) participants had 4 serious adverse events, all considered unrelated to BAX 335. No serious adverse event led to death. No clinical thrombosis, inhibitors, or other FIX Padua-directed immunity was reported. FIX expression was measurable in 7 of 8 participants; peak FIX activity displayed dose dependence (32.0% to 58.5% in cohort 3). One participant achieved sustained therapeutic FIX activity of ∼20%, without bleeding or replacement therapy, for 4 years; in others, FIX activity was not sustained beyond 5 to 11 weeks. In contrast to some previous studies, corticosteroid treatment did not stabilize FIX activity loss. We hypothesize that the loss of transgene expression could have been caused by stimulation of innate immune responses, including CpG oligodeoxynucleotides introduced into the BAX 335 coding sequence by codon optimization. This trial was registered at www.clinicaltrials.gov as #NCT01687608.


Subject(s)
CpG Islands/genetics , Factor IX/therapeutic use , Gene Expression Regulation , Genetic Therapy , Hemophilia B/therapy , Recombinant Fusion Proteins/therapeutic use , Adolescent , Adult , Aged , Chemical and Drug Induced Liver Injury/etiology , Factor IX/biosynthesis , Factor IX/genetics , Gain of Function Mutation , Hemophilia B/genetics , Hemophilia B/immunology , Humans , Immunity, Innate , Male , Middle Aged , Pathogen-Associated Molecular Pattern Molecules/immunology , Prospective Studies , Rhabdomyolysis/etiology , Toll-Like Receptor 9/physiology , Transgenes , Young Adult
2.
Biotechnol Prog ; 36(2): e2938, 2020 03.
Article in English | MEDLINE | ID: mdl-31677255

ABSTRACT

Replacement therapy with recombinant drugs is the main therapeutic strategy for hemophilia B patients. To reduce the production costs of recombinant coagulation factors, improvement of their expression and activity by enhancement of γ-carboxylation might be of interest. The expression and functional activity of vitamin K-dependent (VKD) coagulation proteins rely, in part, on the VKD process of γ-carboxylation that is mediated by the enzymes γ-carboxylase and vitamin K epoxide reductase (VKOR). Since the recombinant production of VKD proteins is hampered by the inefficiency of this enzymatic process, we specifically have examined the stable expression of functional blood coagulation factor IX (FIX) in HEK293 cells following transient overexpression of VKORC1 as an important part of VKOR component. Recombinant hFIX-producing human embryonic kidney (HEK) cells were transfected to overexpress VKORC1. Following reverse transcription polymerase chain reaction (RT-PCR) analysis, expression efficiency of the active hFIX was analyzed by performing enzyme-linked immunosorbent assay and coagulation test. In addition, to quantify γ-carboxylated recombinant FIX, the barium citrate method was used. Overexpression of VKORC1 in FIX-producing HEK cells, resulting in a 3.2-fold higher expression of functional FIX, which displayed a 1.4-fold enhanced specific activity. Moreover, a 3.9-fold enhanced recovery of fully γ-carboxylated FIX following barium citrate adsorption was achieved. Collectively, these findings indicate that the overexpression of VKORC1 results in the production of higher levels of functional hFIX in HEK293 cells. The increase of the VKORC1 as a supplier of γ-carboxylase seems to play a significant role in increasing the amount and efficiency of recombinant FIX production, thereby reducing the production costs.


Subject(s)
Cell Engineering , Factor IX/biosynthesis , Vitamin K Epoxide Reductases/genetics , Cells, Cultured , Factor IX/analysis , HEK293 Cells , Humans , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis
4.
Blood ; 131(9): 1022-1031, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29246900

ABSTRACT

Gene therapy for hemophilia B aims to ameliorate bleeding risk and provide endogenous factor IX (FIX) activity/synthesis through a single treatment, eliminating the requirement for FIX concentrate. AMT-060 combines an adeno-associated virus-5 (AAV5) vector with a liver-specific promoter driving expression of a codon-optimized wild-type human FIX gene. This multinational, open-label study included 10 adults with hemophilia B (FIX ≤2% of normal) and severe-bleeding phenotype. No participants tested positive for AAV5-neutralizing antibodies using a green-fluorescent protein-based assay, and all 10 were enrolled. A single dose of 5 × 1012 or 2 × 1013 genome copies of AMT-060/kilogram was administered to 5 participants each. In the low-dose cohort, mean endogenous FIX activity increased to 4.4 IU/dL. Annualized FIX use was reduced by 81%, and mean annualized spontaneous bleeding rate (ASBR) decreased from 9.8% to 4.6% (53%). In the higher-dose cohort, mean FIX activity increased to 6.9 IU/dL. Annualized FIX use decreased by 73%, and mean ASBR declined from 3.0 to 0.9 (70%). There was no reduction in traumatic bleeds. FIX activity was stable in both cohorts, and 8 of 9 participants receiving FIX at study entry stopped prophylaxis. Limited, asymptomatic, and transient alanine aminotransferase elevations in the low-dose (n = 1) and higher-dose (n = 2) cohorts were treated with prednisolone. No decrease in FIX activity or capsid-specific T-cell responses were detected during transaminase elevations. A single infusion of AMT-060 had a positive safety profile and resulted in stable and clinically important increases in FIX activity, a marked reduction in spontaneous bleeds and FIX concentrate use, without detectable cellular immune responses against capsids. This trial was registered at www.clinicaltrials.gov as #NCT02396342; EudraCT #2013-005579-42.


Subject(s)
Factor IX , Genetic Therapy , Genetic Vectors , Hemophilia B , Parvovirinae , Prednisolone/administration & dosage , Adult , Dependovirus , Factor IX/biosynthesis , Factor IX/genetics , Female , Hemophilia B/blood , Hemophilia B/genetics , Hemophilia B/therapy , Humans , Male
5.
Hematology Am Soc Hematol Educ Program ; 2017(1): 587-594, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29222308

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.


Subject(s)
Dependovirus , Factor IX , Factor VIII , Genetic Therapy/methods , Hemophilia A , Transduction, Genetic/methods , Animals , Factor IX/biosynthesis , Factor IX/genetics , Factor VIII/biosynthesis , Factor VIII/genetics , Hemophilia A/genetics , Hemophilia A/metabolism , Hemophilia A/therapy , Humans
6.
Bioengineered ; 8(5): 462-470, 2017 Sep 03.
Article in English | MEDLINE | ID: mdl-28277160

ABSTRACT

The main treatment option for Hemophilia A/B patients involves the administration of recombinant coagulation factors on-demand or in a prophylactic approach. Despite the safety and efficacy of this replacement therapy, the development of antibodies against the coagulation factor infused, which neutralize the procoagulant activity, is a severe complication. The production of recombinant coagulation factors in human cell lines is an efficient approach to avoid such complication. Human cell lines can produce recombinant proteins with post translation modifications more similar to their natural counterpart, reducing potential immunogenic reactions. This review provides a brief overview of the most important characteristics of recombinant FVIII and FIX products available on the market and the improvements that have recently been achieved by the production using human cell lines.


Subject(s)
Factor IX/biosynthesis , Factor IX/genetics , Factor VIII/biosynthesis , Factor VIII/genetics , Genetic Enhancement/methods , Protein Engineering/methods , Animals , Blood Coagulation Factors/biosynthesis , Blood Coagulation Factors/genetics , COS Cells , Cloning, Molecular/methods , HEK293 Cells , Hep G2 Cells , Humans , Protein Conformation , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Species Specificity
7.
Hum Gene Ther Methods ; 27(3): 112-21, 2016 06.
Article in English | MEDLINE | ID: mdl-27229773

ABSTRACT

Adeno-associated virus (AAV) vectors are increasingly popular in gene therapy because they are unassociated with human disease, replication dependent, and less immunogenic than other viral vectors and can infect a variety of cell types. These vectors have been used in over 130 clinical trials, and one AAV product has been approved for treatment of lipoprotein lipase deficiency in Europe. To meet the demand for the increasing quantities of AAV required for clinical trials and treatment, a scalable high-capacity technology is required. Bioreactors meet these requirements but limited options are available for adherent HEK 293T/17 cells. Here we optimize the transient transfection of HEK293T/17 cells for the production of AAV human factor IX in a disposable fixed-bed bioreactor, the iCELLis(®) Nano (PALL Corporation). A fixed bed in the center of the iCELLis bioreactor is surrounded by culture medium that is pumped through the bed from the bottom of the bioreactor so that a thin film of the medium overflows the bed and is replenished with oxygen and depleted of CO2 as it returns to the surrounding medium reservoir. We show that this fixed-bed bioreactor can support as many as 2.5 × 10(8) cells/ml of fixed bed (1.9 × 10(6) cells/cm(2)). By optimizing culture and transfection parameters such as the concentration of DNA for transfection, day of harvest, size of PEI/DNA particles, and transfection medium, and adding an additional medium change to the process, we increased our yield to as high as 9.0 × 10(14) viral particles per square meter of fixed bed. We also show an average GFP transfection of 97% of cells throughout the fixed bed. These yields make the iCELLis a promising scalable technology for the clinical production of AAV gene therapy products.


Subject(s)
Factor IX/biosynthesis , Genetic Therapy , Genetic Vectors/therapeutic use , Bioreactors , Dependovirus/genetics , Factor IX/genetics , HEK293 Cells , Humans , Transfection
8.
Protein Expr Purif ; 121: 149-56, 2016 May.
Article in English | MEDLINE | ID: mdl-26802680

ABSTRACT

Factor IX (FIX) is a vitamin K-dependent protein, and it has become a valuable pharmaceutical in the Hemophilia B treatment. We evaluated the potential of recombinant human FIX (rhFIX) expression in 293T and SK-Hep-1 human cell lines. SK-Hep-1-FIX cells produced higher levels of biologically active protein. The growth profile of 293T-FIX cells was not influenced by lentiviral integration number into the cellular genome. SK-Hep-1-FIX cells showed a significantly lower growth rate than SK-Hep-1 cells. γ-carboxylation process is significant to FIX biological activity, thus we performed a expression analysis of genes involved in this process. The 293T gene expression suggests that this cell line could efficiently carboxylate FIX, however only 28% of the total secreted protein is active. SK-Hep-1 cells did not express high amounts of VKORC1 and carboxylase, but this cell line secreted large amounts of active protein. Enrichment of culture medium with Ca(+2) and Mg(+2) ions did not affect positively rhFIX expression in SK-Hep-1 cells. In 293T cells, the addition of 0.5 mM Ca(+2) and 1 mM Mg(+2) resulted in higher rhFIX concentration. SK-Hep-1 cell line proved to be very effective in rhFIX production, and it can be used as a novel biotechnological platform for the production of recombinant proteins.


Subject(s)
Biotechnology , Factor IX/biosynthesis , Recombinant Proteins/biosynthesis , Factor IX/genetics , Gene Expression , Genetic Vectors , HEK293 Cells , Humans , Recombinant Proteins/genetics
9.
J Thromb Haemost ; 13 Suppl 1: S151-60, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26149016

ABSTRACT

Factor IX deficiency (hemophilia B) is less common than factor VIII deficiency (hemophilia A), and innovations in therapy for hemophilia B have generally lagged behind those for hemophilia A. Recently, the first sustained correction of the hemophilia bleeding phenotype by clotting factor gene therapy has been described using recombinant adeno-associated virus (AAV) to deliver factor IX. Despite this success, many individuals with hemophilia B, including children, men with active hepatitis, and individuals who have pre-existing natural immunity to AAV, are not eligible for the current iteration of hemophilia B gene therapy. In addition, recent advances in recombinant factor IX protein engineering have led some hemophilia treaters to reconsider the urgency of genetic cure. Current clinical and preclinical approaches to advancing AAV-based and alternative approaches to factor IX gene therapy are considered in the context of current demographics and treatment of the hemophilia B population.


Subject(s)
Genetic Therapy/methods , Hemophilia B/therapy , Animals , Dependovirus/genetics , Factor IX/biosynthesis , Factor IX/genetics , Factor IX/therapeutic use , Gene Transfer Techniques , Genetic Predisposition to Disease , Genetic Vectors , Hemophilia B/blood , Hemophilia B/diagnosis , Hemophilia B/genetics , Hemostatics/therapeutic use , Humans , Lentivirus/genetics , Male , Recombinant Proteins/therapeutic use , Treatment Outcome
10.
J Thromb Haemost ; 13 Suppl 1: S161-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26149017

ABSTRACT

Hemophilia remains a non-curative disease, and patients are constrained to undergo repeated injections of clotting factors. In contrast, the sustained production of endogenous factors VIII (FVIII) or IX (FIX) by the patient's own cells could represent a curative treatment. Gene therapy has thus provided new hope for these patients. However, the issues surrounding the durability of expression and immune responses against gene transfer vectors remain. Cell therapy, involving stem cells expanded in vitro, can provide de novo protein synthesis and, if implanted successfully, could induce a steady-state production of low quantities of factors, which may keep the patient above the level required to prevent spontaneous bleeding. Liver-derived stem cells are already being assessed in clinical trials for inborn errors of metabolism and, in view of their capacity to produce FVIII and FIX in cell culture, they are now also being considered for clinical application in hemophilia patients.


Subject(s)
Factor IX/biosynthesis , Factor VIII/biosynthesis , Genetic Therapy/methods , Hemophilia A/surgery , Hemorrhage/prevention & control , Liver/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Animals , Extracellular Vesicles/metabolism , Factor IX/genetics , Factor VIII/genetics , Hemophilia A/blood , Hemophilia A/diagnosis , Hemophilia A/genetics , Hemostasis , Humans , Treatment Outcome
11.
J Biotechnol ; 213: 20-7, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-25738489

ABSTRACT

Recombinant clotting factors are important biotherapeutics that Pfizer has produced and marketed for over fifteen years. Owing to the complexity of the structure and function of these blood factors, it can be challenging to achieve the required product quality and manufacturing productivity. The article highlights the semi-continuous and continuous cell culture processes employed by Pfizer for the production of BeneFIX and ReFacto AF. The benefits of such processes, the challenges of maintaining an aseptic production culture for extended periods, and batch definition are discussed in this article.


Subject(s)
Bioreactors , Cell Culture Techniques , Factor IX/biosynthesis , Factor VIII/biosynthesis , Animals , CHO Cells , Cricetulus
13.
Sci Rep ; 4: 7403, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25492703

ABSTRACT

Transgene insertion plays an important role in gene therapy and in biological studies. Transposon-based systems that integrate transgenes by transposase-catalyzed "cut-and-paste" mechanism have emerged as an attractive system for transgenesis. Hyperactive piggyBac transposon is particularly promising due to its ability to integrate large transgenes with high efficiency. However, prolonged expression of transposase can become a potential source of genotoxic effects due to uncontrolled transposition of the integrated transgene from one chromosomal locus to another. In this study we propose a vector design to decrease post-transposition expression of transposase and to eliminate the cells that have residual transposase expression. We design a single plasmid construct that combines the transposase and the transpositioning transgene element to share a single polyA sequence for termination. Consequently, the separation of the transposase element from the polyA sequence after transposition leads to its deactivation. We also co-express Herpes Simplex Virus thymidine kinase (HSV-tk) with the transposase. Therefore, cells having residual transposase expression can be eliminated by the administration of ganciclovir. We demonstrate the utility of this combination transposon system by integrating and expressing a model therapeutic gene, human coagulation Factor IX, in HEK293T cells.


Subject(s)
DNA Transposable Elements , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Factor IX/biosynthesis , Factor IX/genetics , HEK293 Cells , Humans , Thymidine Kinase/biosynthesis , Thymidine Kinase/genetics , Transposases/biosynthesis , Transposases/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
14.
Clin Lab ; 60(8): 1325-31, 2014.
Article in English | MEDLINE | ID: mdl-25185418

ABSTRACT

BACKGROUND: The goal of this study was to compare the validity of two laboratory assays, rotation thromboelastometry (ROTEM) and endogenous thrombin potential (ETP), in monitoring and evaluating different prophylactic treatment regimens in patients with severe haemophilia. METHODS: Twenty adult patients with severe haemophilia were divided into three groups according to treatment regimen with concentrate of factor (F) VIII/IX: full-dose prophylaxis (5 patients), intermediate-dose prophylaxis (5 patients), and on demand treatment (10 patients). RESULTS: The ROTEM for the group treated with full-dose prophylaxis was significantly lower than ROTEM for the group treated with intermediate-dose prophylaxis (p = 0.025). Among the patients given full-dose prophylaxis, 40% (2 patients) had prolonged ROTEM after 3 months of treatment, while among those given intermediate-dose prophylaxis all patients (100%, 5 patients) had prolonged ROTEM (p = 0.038). The ETP was significantly improved after 3 months of full-dose in comparison with intermediate-dose prophylaxis (p = 0.042). CONCLUSIONS: ROTEM and ETP are useful laboratory assays for monitoring efficacy of different prophylaxis regimens with concentrate of FVIII/IX in patients with severe haemophilia, helping in making decisions regarding optimal dose-regimen prophylaxis.


Subject(s)
Hemophilia A/blood , Hemophilia A/prevention & control , Adult , Blood Coagulation Tests , Case-Control Studies , Clinical Laboratory Techniques , Drug Monitoring/methods , Factor IX/biosynthesis , Factor VIII/biosynthesis , Humans , Male , Middle Aged , Reproducibility of Results , Thrombelastography/methods , Thrombin/biosynthesis , Thrombin/chemistry , Time Factors , Young Adult
15.
J Thromb Haemost ; 12(11): 1861-73, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25224783

ABSTRACT

BACKGROUND: FVIII neutralizing antibodies are the main complication of substitution therapy in hemophilia A (HA); auto-antibodies against FVIII causing acquired HA can also occur. Treatment of inhibitor patients remains challenging because prophylactic treatment with existing FVIII bypassing agents, all based on constitutively active coagulation factors, is difficult due to their short half-life. OBJECTIVES: To generate zymogenic FIX variants with FVIII-independent activity for gene- and protein-based therapy for HA. METHODS: Modifications were introduced into FIX based on current knowledge of FIX structure and FVIII-independent function followed by random screening. Activity, thrombin generation and FX activation by FIX mutants were characterized in the presence and absence of FVIII. Phenotype correction of promising candidates was assessed by the tail-clip assay in FVIII-knockout mice. RESULTS: About 1600 clones were screened and three mutations (L6F, S102N and E185D) identified, which improved FVIII-independent activity in combination with our previously described variant FIX-ITV. By systematic combination of all mutations, six FIX mutants with the desired bypassing activity were designed. Candidate mutants FIX-IDAV and FIX-FIAV demonstrated the most efficient thrombin generation in FVIII-deficient plasma and had considerably increased activities towards FX in the absence of FVIII, in that they showed an up to 5-fold increase in catalytic efficiency. Expression of FIX-IDAV in FVIII knockout mice reduced blood loss after the tail-clip assay, even in the presence of neutralizing FVIII antibodies. CONCLUSION: Activatable bioengineered FIX molecules (as opposed to pre-activated coagulation factors) with FVIII-independent activity might be a promising tool for improving HA treatment, especially for patients with inhibitors.


Subject(s)
Factor IX/genetics , Factor VIII/metabolism , Genetic Therapy/methods , Hemophilia A/therapy , Hemostasis , Mutation , Animals , Dependovirus/genetics , Disease Models, Animal , Factor IX/biosynthesis , Factor VIII/biosynthesis , Factor VIII/genetics , Factor Xa/metabolism , Gene Transfer Techniques , Genetic Vectors , HEK293 Cells , Hemophilia A/blood , Hemophilia A/diagnosis , Humans , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Thrombin/metabolism , Time Factors , Transfection
16.
Sheng Wu Gong Cheng Xue Bao ; 30(3): 492-503, 2014 Mar.
Article in Chinese | MEDLINE | ID: mdl-25007585

ABSTRACT

We established methods to isolate human amniotic fluid-derived progenitor cells (hAFPCs), and analyze the ability of hAFPCs to secrete human coagulation factor IX (hFIX) after gene modification. The hAFPCs were manually isolated by selection for attachment to gelatin coated culture dish. hFIX cDNA was transfected into hAPFCs by using a lentiviral vector. The hFIX protein concentration and activity produced from hAFPCs were determined by enzyme-linked immunosorbent assay (ELISA) and clotting assay. The isolated spindle-shaped cells showed fibroblastoid morphology after three culture passages. The doubling time in culture was 39.05 hours. Immunocytochemistry staining of the fibroblast-like cells from amniotic fluid detected expression of stem cell markers such as SSEA4 and TRA1-60. Quantitative PCR analysis demonstrated the expression of NANOG, OCT4 and SOX2 mRNAs. Transfected hAFPCs could produce and secrete hFIX into the culture medium. The observed concentration of secreted hFIX was 20.37% +/- 2.77% two days after passage, with clotting activity of 16.42% +/- 1.78%. The amount of hFIX:Ag reached a plateau of 50.35% +/- 5.42%, with clotting activity 45.34% +/- 4.67%. In conclusion, this study established method to isolate and culture amniotic fluid progenitor cells. Transfected hAFPCs can produce hFIX at stable levels in vitro, and clotting activity increases with higher hFIX concentration. Genetically engineered hAFPC are a potential method for prenatal treatment of hemophilia B.


Subject(s)
Amniotic Fluid/cytology , Cell Separation/methods , Factor IX/biosynthesis , Genetic Engineering , Stem Cells/cytology , Blood Coagulation , Cell Culture Techniques , DNA, Complementary , Genetic Vectors , Humans , Stem Cells/metabolism , Transfection
17.
Biomed Res Int ; 2014: 315375, 2014.
Article in English | MEDLINE | ID: mdl-24955355

ABSTRACT

Production of biopharmaceuticals from transgenic animal milk is a cost-effective method for highly complex proteins that cannot be efficiently produced using conventional systems such as microorganisms or animal cells. Yields of recombinant human factor IX (rhFIX) produced from transgenic porcine milk under the control of the bovine α-lactalbumin promoter reached 0.25 mg/mL. The rhFIX protein was purified from transgenic porcine milk using a three-column purification scheme after a precipitation step to remove casein. The purified protein had high specific activity and a low ratio of the active form (FIXa). The purified rhFIX had 11.9 γ-carboxyglutamic acid (Gla) residues/mol protein, which approached full occupancy of the 12 potential sites in the Gla domain. The rhFIX was shown to have a higher isoelectric point and lower sialic acid content than plasma-derived FIX (pdFIX). The rhFIX had the same N-glycosylation sites and phosphorylation sites as pdFIX, but had a higher specific activity. These results suggest that rhFIX produced from porcine milk is physiologically active and they support the use of transgenic animals as bioreactors for industrial scale production in milk.


Subject(s)
Factor IX/biosynthesis , Milk/chemistry , Recombinant Proteins/biosynthesis , Animals , Animals, Genetically Modified , Bioreactors , Cattle , Factor IX/genetics , Humans , Milk/metabolism , Recombinant Proteins/genetics , Swine
18.
J Thromb Haemost ; 12(6): 932-42, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24679056

ABSTRACT

BACKGROUND: Oral gene delivery of non-viral vectors is an attractive strategy to achieve transgene expression. Although expected efficacy from non-viral delivery systems is relatively low, repeated vector administration is possible and may help to obtain durable transgene expression in a therapeutic range. OBJECTIVES: To test the principle feasibility of using factor (F) IX variants with improved function combined with an optimized oral delivery system in hemophilia B (HB) mice. METHODS: FIX modifications were introduced by site-directed mutagenesis into plasmid- or minicircle-based expression cassettes. Vectors were formulated as chitosan nanoparticles for oral delivery to HB mice. Protection of vector DNA in nanoparticle constructs and transfection efficiency were characterized. HB mice received eGFP-formulated chitosan nanoparticles to confirm gene transfer in vivo. FIX expression, phenotype correction and the potential of nanoparticles to induce immunotolerance (ITI) against exogenous FIX were evaluated after repeated oral administration. RESULTS: Transfection of HEK 293T cells or livers of FIX-knockout mice with nanoparticles resulted in GFP or functional FIX expression. Oral administration of FIX mutants resulted in exclusive FIX expression in the small intestine, as confirmed by RT-PCR and fluorescence staining. HB mice demonstrated transient FIX expression reaching > 14% of normal activity and partial phenotype correction after oral delivery of FIX mutants with high specific activity and improved tissue release. CONCLUSION: The feasibility of oral, non-viral delivery of FIX was established and improved by bioengineered FIX proteins and optimized vectors. Thus, these data might point the way for development of a clinically applicable oral gene transfer strategy for hemophilia B.


Subject(s)
Chitosan/administration & dosage , Factor IX/administration & dosage , Factor IX/genetics , Genetic Therapy/methods , Hemophilia B/therapy , Mutation , Administration, Oral , Animals , Disease Models, Animal , Factor IX/biosynthesis , Feasibility Studies , Genotype , HEK293 Cells , Hemophilia B/blood , Hemophilia B/genetics , Hemostasis , Humans , Intestine, Small/metabolism , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Nanoparticles , Phenotype , Time Factors , Transfection
19.
Blood ; 122(19): 3283-7, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24085764

ABSTRACT

Monogenic diseases, including hemophilia, represent ideal targets for genome-editing approaches aimed at correcting a defective gene. Here we report that systemic adeno-associated virus (AAV) vector delivery of zinc finger nucleases (ZFNs) and corrective donor template to the predominantly quiescent livers of adult mice enables production of high levels of human factor IX in a murine model of hemophilia B. Further, we show that off-target cleavage can be substantially reduced while maintaining robust editing by using obligate heterodimeric ZFNs engineered to minimize unwanted cleavage attributable to homodimerization of the ZFNs. These results broaden the therapeutic potential of AAV/ZFN-mediated genome editing in the liver and could expand this strategy to other nonreplicating cell types.


Subject(s)
Endonucleases/genetics , Factor IX/biosynthesis , Genetic Therapy/methods , Genetic Vectors , Genome , Hemophilia B/therapy , Zinc Fingers/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Endonucleases/metabolism , Factor IX/genetics , Factor IX/metabolism , Hemophilia B/genetics , Hemophilia B/pathology , Liver/metabolism , Male , Mice , Mice, Transgenic , Protein Multimerization
20.
PLoS One ; 8(8): e73423, 2013.
Article in English | MEDLINE | ID: mdl-24015305

ABSTRACT

Topical gene delivery to the epidermis has the potential to be an effective therapy for skin disorders, cutaneous cancers, vaccinations and systemic metabolic diseases. Previously, we reported on a non-invasive multielectrode array (MEA) that efficiently delivered plasmid DNA and enhanced expression to the skin of several animal models by in vivo gene electrotransfer. Here, we characterized plasmid DNA delivery with the MEA in a hairless guinea pig model, which has a similar histology and structure to human skin. Significant elevation of gene expression up to 4 logs was achieved with intradermal DNA administration followed by topical non-invasive skin gene electrotransfer. This delivery produced gene expression in the skin of hairless guinea pig up to 12 to 15 days. Gene expression was observed exclusively in the epidermis. Skin gene electrotransfer with the MEA resulted in only minimal and mild skin changes. A low level of human Factor IX was detected in the plasma of hairless guinea pig after gene electrotransfer with the MEA, although a significant increase of Factor IX was obtained in the skin of animals. These results suggest gene electrotransfer with the MEA can be a safe, efficient, non-invasive skin delivery method for skin disorders, vaccinations and potential systemic diseases where low levels of gene products are sufficient.


Subject(s)
Electroporation/methods , Epidermis/metabolism , Gene Transfer Techniques , Plasmids/pharmacology , Skin Neoplasms/therapy , Animals , Epidermis/pathology , Factor IX/biosynthesis , Factor IX/genetics , Female , Guinea Pigs , Humans , Skin Neoplasms/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...