Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 13(1): 2454, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35508619

ABSTRACT

Liver gene therapy with adeno-associated viral (AAV) vectors delivering clotting factor transgenes into hepatocytes has shown multiyear therapeutic benefit in adults with hemophilia. However, the mostly episomal nature of AAV vectors challenges their application to young pediatric patients. We developed lentiviral vectors, which integrate in the host cell genome, that achieve efficient liver gene transfer in mice, dogs and non-human primates, by intravenous delivery. Here we first compare engineered coagulation factor VIII transgenes and show that codon-usage optimization improved expression 10-20-fold in hemophilia A mice and that inclusion of an unstructured XTEN peptide, known to increase the half-life of the payload protein, provided an additional >10-fold increase in overall factor VIII output in mice and non-human primates. Stable nearly life-long normal and above-normal factor VIII activity was achieved in hemophilia A mouse models. Overall, we show long-term factor VIII activity and restoration of hemostasis, by lentiviral gene therapy to hemophilia A mice and normal-range factor VIII activity in non-human primate, paving the way for potential clinical application.


Subject(s)
Hemophilia A , Animals , Child , Dogs , Factor VIII/genetics , Genetic Therapy , Genetic Vectors/genetics , Hemophilia A/genetics , Hemophilia A/therapy , Humans , Liver/metabolism , Mice , Primates/genetics
2.
Blood ; 135(17): 1484-1496, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32078672

ABSTRACT

Factor VIII (FVIII) replacement products enable comprehensive care in hemophilia A. Treatment goals in severe hemophilia A are expanding beyond low annualized bleed rates to include long-term outcomes associated with high sustained FVIII levels. Endogenous von Willebrand factor (VWF) stabilizes and protects FVIII from degradation and clearance, but it also subjects FVIII to a half-life ceiling of ∼15 to 19 hours. Increasing recombinant FVIII (rFVIII) half-life further is ultimately dependent upon uncoupling rFVIII from endogenous VWF. We have developed a new class of FVIII replacement, rFVIIIFc-VWF-XTEN (BIVV001), that is physically decoupled from endogenous VWF and has enhanced pharmacokinetic properties compared with all previous FVIII products. BIVV001 was bioengineered as a unique fusion protein consisting of a VWF-D'D3 domain fused to rFVIII via immunoglobulin-G1 Fc domains and 2 XTEN polypeptides (Amunix Pharmaceuticals, Inc, Mountain View, CA). Plasma FVIII half-life after BIVV001 administration in mice and monkeys was 25 to 31 hours and 33 to 34 hours, respectively, representing a three- to fourfold increase in FVIII half-life. Our results showed that multifaceted protein engineering, far beyond a few amino acid substitutions, could significantly improve rFVIII pharmacokinetic properties while maintaining hemostatic function. BIVV001 is the first rFVIII with the potential to significantly change the treatment paradigm for severe hemophilia A by providing optimal protection against all bleed types, with less frequent doses. The protein engineering methods described herein can also be applied to other complex proteins.


Subject(s)
Factor VIII/metabolism , Hemophilia A/therapy , Hemorrhage/prevention & control , Recombinant Fusion Proteins/administration & dosage , von Willebrand Factor/metabolism , Animals , Factor VIII/genetics , Hemophilia A/metabolism , Hemophilia A/pathology , Hemostasis , Humans , Male , Mice , Mice, Inbred C57BL , Primates , von Willebrand Factor/genetics
3.
Sci Transl Med ; 11(493)2019 05 22.
Article in English | MEDLINE | ID: mdl-31118293

ABSTRACT

Liver-directed gene therapy for the coagulation disorder hemophilia showed safe and effective results in clinical trials using adeno-associated viral vectors to replace a functional coagulation factor, although some unmet needs remain. Lentiviral vectors (LVs) may address some of these hurdles because of their potential for stable expression and the low prevalence of preexisting viral immunity in humans. However, systemic LV administration to hemophilic dogs was associated to mild acute toxicity and low efficacy at the administered doses. Here, exploiting intravital microscopy and LV surface engineering, we report a major role of the human phagocytosis inhibitor CD47, incorporated into LV cell membrane, in protecting LVs from uptake by professional phagocytes and innate immune sensing, thus favoring biodistribution to hepatocytes after systemic administration. By enforcing high CD47 surface content, we generated phagocytosis-shielded LVs which, upon intravenous administration to nonhuman primates, showed selective liver and spleen targeting and enhanced hepatocyte gene transfer compared to parental LV, reaching supraphysiological activity of human coagulation factor IX, the protein encoded by the transgene, without signs of toxicity or clonal expansion of transduced cells.


Subject(s)
Genetic Therapy , Genetic Vectors/therapeutic use , Lentivirus/genetics , Liver/pathology , Phagocytosis , Animals , CD47 Antigen/metabolism , Gene Transfer Techniques , Hepatocytes/metabolism , Humans , Immune Tolerance , Immunity, Innate , Kupffer Cells/metabolism , Macaca , Mice, Inbred C57BL , Mice, Inbred NOD , Phagocytes/metabolism , Tissue Distribution
4.
Cell Immunol ; 301: 30-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26775174

ABSTRACT

Anti-factor VIII (FVIII) antibodies is a major complication of FVIII replacement therapy for hemophilia A. We investigated the immune response to recombinant human factor VIII Fc (rFVIIIFc) in comparison to BDD-rFVIII and full-length rFVIII (FL-rFVIII) in hemophilia A mice. Repeated administration of therapeutically relevant doses of rFVIIIFc in these mice resulted in significantly lower antibody responses to rFVIII compared to BDD-rFVIII and FL-rFVIII and reduced antibody production upon subsequent challenge with high doses of rFVIIIFc. The induction of a tolerogenic response by rFVIIIFc was associated with higher percentage of regulatory T-cells, a lower percentage of pro-inflammatory splenic T-cells, and up-regulation of tolerogenic cytokines and markers. Disruption of Fc interactions with either FcRn or Fcγ receptors diminished tolerance induction, suggesting the involvement of these pathways. These results indicate that rFVIIIFc reduces immunogenicity and imparts tolerance to rFVIII demonstrating that recombinant therapeutic proteins may be modified to influence immunogenicity and facilitate tolerance.


Subject(s)
Factor VIII/immunology , Hemophilia A/immunology , Immune Tolerance/immunology , Immunoglobulin Fc Fragments/immunology , Recombinant Fusion Proteins/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Factor VIII/pharmacology , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/pharmacology , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/immunology
5.
PLoS One ; 10(4): e0124930, 2015.
Article in English | MEDLINE | ID: mdl-25905473

ABSTRACT

We recently developed a longer lasting recombinant factor VIII-Fc fusion protein, rFVIIIFc, to extend the half-life of replacement FVIII for the treatment of people with hemophilia A. In order to elucidate the biological mechanism for the elongated half-life of rFVIIIFc at a cellular level we delineated the roles of VWF and the tissue-specific expression of the neonatal Fc receptor (FcRn) in the biodistribution, clearance and cycling of rFVIIIFc. We find the tissue biodistribution is similar for rFVIIIFc and rFVIII and that liver is the major clearance organ for both molecules. VWF reduces the clearance and the initial liver uptake of rFVIIIFc. Pharmacokinetic studies in FcRn chimeric mice show that FcRn expressed in somatic cells (hepatocytes or liver sinusoidal endothelial cells) mediates the decreased clearance of rFVIIIFc, but FcRn in hematopoietic cells (Kupffer cells) does not affect clearance. Immunohistochemical studies show that when rFVIII or rFVIIIFc is in dynamic equilibrium binding with VWF, they mostly co localize with VWF in Kupffer cells and macrophages, confirming a major role for liver macrophages in the internalization and clearance of the VWF-FVIII complex. In the absence of VWF a clear difference in cellular localization of VWF-free rFVIII and rFVIIIFc is observed and neither molecule is detected in Kupffer cells. Instead, rFVIII is observed in hepatocytes, indicating that free rFVIII is cleared by hepatocytes, while rFVIIIFc is observed as a diffuse liver sinusoidal staining, suggesting recycling of free-rFVIIIFc out of hepatocytes. These studies reveal two parallel linked clearance pathways, with a dominant pathway in which both rFVIIIFc and rFVIII complexed with VWF are cleared mainly by Kupffer cells without FcRn cycling. In contrast, the free fraction of rFVIII or rFVIIIFc unbound by VWF enters hepatocytes, where FcRn reduces the degradation and clearance of rFVIIIFc relative to rFVIII by cycling rFVIIIFc back to the liver sinusoid and into circulation, enabling the elongated half-life of rFVIIIFc.


Subject(s)
Factor VIII/metabolism , Hepatocytes/metabolism , Histocompatibility Antigens Class I/physiology , Receptors, Fc/physiology , von Willebrand Factor/metabolism , Animals , Mice , Mice, Knockout , Receptors, Scavenger/metabolism , Recombinant Fusion Proteins/metabolism , Tissue Distribution
6.
Blood ; 119(13): 3024-30, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22246033

ABSTRACT

Despite proven benefits, prophylactic treatment for hemophilia A is hampered by the short half-life of factor VIII. A recombinant factor VIII-Fc fusion protein (rFVIIIFc) was constructed to determine the potential for reduced frequency of dosing. rFVIIIFc has an ∼ 2-fold longer half-life than rFVIII in hemophilia A (HemA) mice and dogs. The extension of rFVIIIFc half-life requires interaction of Fc with the neonatal Fc receptor (FcRn). In FcRn knockout mice, the extension of rFVIIIFc half-life is abrogated, and is restored in human FcRn transgenic mice. The Fc fusion has no impact on FVIII-specific activity. rFVIIIFc has comparable acute efficacy as rFVIII in treating tail clip injury in HemA mice, and fully corrects whole blood clotting time (WBCT) in HemA dogs immediately after dosing. Furthermore, consistent with prolonged half-life, rFVIIIFc shows 2-fold longer prophylactic efficacy in protecting HemA mice from tail vein transection bleeding induced 24-48 hours after dosing. In HemA dogs, rFVIIIFc also sustains partial correction of WBCT 1.5- to 2-fold longer than rFVIII. rFVIIIFc was well tolerated in both species. Thus, the rescue of FVIII by Fc fusion to provide prolonged protection presents a novel pathway for FVIII catabolism, and warrants further investigation.


Subject(s)
Factor VIII/pharmacokinetics , Hemophilia A/metabolism , Histocompatibility Antigens Class I/pharmacology , Recombinant Fusion Proteins/pharmacokinetics , Animals , Coagulants/pharmacokinetics , Coagulants/therapeutic use , Disease Models, Animal , Dog Diseases/drug therapy , Dog Diseases/metabolism , Dogs , Factor VIII/chemistry , Factor VIII/genetics , Factor VIII/therapeutic use , HEK293 Cells , Half-Life , Hemophilia A/drug therapy , Hemophilia A/pathology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Fc/chemistry , Receptors, Fc/metabolism , Receptors, Fc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Whole Blood Coagulation Time
SELECTION OF CITATIONS
SEARCH DETAIL
...