Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 3: 15053, 2016.
Article in English | MEDLINE | ID: mdl-26858964

ABSTRACT

Enzyme and gene replacement strategies have developed into viable therapeutic approaches for the treatment of Pompe disease (acid α-glucosidase (GAA) deficiency). Unfortunately, the introduction of GAA and viral vectors encoding the enzyme can lead to detrimental immune responses that attenuate treatment benefits and can impact patient safety. Preclinical and clinical experience in addressing humoral responses toward enzyme and gene therapy for Pompe disease have provided greater understanding of the immunological consequences of the provided therapy. B- and T-cell modulation has been shown to be effective in preventing infusion-associated reactions during enzyme replacement therapy in patients and has shown similar success in the context of gene therapy. Additional techniques to induce humoral tolerance for Pompe disease have been the targeted expression or delivery of GAA to discrete cell types or tissues such as the gut-associated lymphoid tissues, red blood cells, hematopoietic stem cells, and the liver. Research into overcoming preexisting immunity through immunomodulation and gene transfer are becoming increasingly important to achieve long-term efficacy. This review highlights the advances in therapies as well as the improved understanding of the molecular mechanisms involved in the humoral immune response with emphasis on methods employed to overcome responses associated with enzyme and gene therapies for Pompe disease.

2.
Hum Gene Ther ; 27(1): 43-59, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26603344

ABSTRACT

Pompe disease is a progressive neuromuscular disorder caused by lysosomal accumulation of glycogen from a deficiency in acid alpha-glucosidase (GAA). Replacement of the missing enzyme is available by repeated protein infusions; however, efficacy is limited by immune response and inability to restore enzymatic function in the central nervous system. An alternative therapeutic option is adeno-associated virus (AAV)-mediated gene therapy, which results in widespread gene transfer and prolonged transgene expression. Both enzyme replacement therapy (ERT) and gene therapy can elicit anti-GAA immune reactions that dampen their effectiveness and pose life-threatening risks to patient safety. To modulate the immune responses related to gene therapy, we show that a human codon-optimized GAA (coGAA) driven by a liver-specific promoter (LSP) using AAV9 is capable of promoting immune tolerance in a Gaa(-/-) mouse model. Copackaging AAV9-LSP-coGAA with the tissue-restricted desmin promoter (AAV9-DES-coGAA) demonstrates the necessary cell autonomous expression in cardiac muscle, skeletal muscle, peripheral nerve, and the spinal cord. Simultaneous high-level expression in liver led to the expansion of GAA-specific regulatory T-cells (Tregs) and induction of immune tolerance. Transfer of Tregs into naïve recipients prevented pathogenic allergic reactions after repeated ERT challenges. Copackaged AAV9 also attenuated preexisting humoral and cellular immune responses, which enhanced the biochemical correction. Our data present a therapeutic design in which simultaneous administration of two copackaged AAV constructs may provide therapeutic benefit and resolve immune reactions in the treatment of multisystem disorders.


Subject(s)
Genetic Therapy , Glucan 1,4-alpha-Glucosidase/genetics , Glycogen Storage Disease Type II/genetics , Immune Tolerance/genetics , Animals , Dependovirus/genetics , Dependovirus/immunology , Glucan 1,4-alpha-Glucosidase/deficiency , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/therapy , Humans , Mice , Mice, Knockout , Promoter Regions, Genetic , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
3.
Clin Immunol ; 158(2): 140-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25842186

ABSTRACT

Antibodies formed against the therapeutic protein are a life-threatening complication that arises during enzyme replacement therapy for Pompe disease (acid α-glucosidase deficiency; GAA). To provide an effective alternative to current practices, we investigated the capacity of anti-B-cell activating factor (BAFF) as a novel drug candidate to prevent antibody formation in a Pompe disease mouse model. A BAFF-neutralizing antibody was administered prophylactically and with maintenance doses in association with enzyme replacement therapy using recombinant human GAA in Gaa(-/-) mice. BAFF blockade delayed antibody production and increased GAA activity within tissues with protection from anaphylaxis. Anti-BAFF also resolved antibody formation during an immune response and precluded the maturation of antibody secreting cells from entering the bone marrow compartment. This treatment modality may therefore be a viable alternative for the clinical management of antibody formation for Pompe disease and has potential use against antibody formation in other protein replacement therapies.


Subject(s)
Antibodies/immunology , B-Cell Activating Factor/antagonists & inhibitors , Enzyme Replacement Therapy , Glucan 1,4-alpha-Glucosidase/therapeutic use , Glycogen Storage Disease Type II/immunology , Animals , Disease Models, Animal , Female , Gene Deletion , Gene Expression Regulation, Enzymologic , Genetic Therapy/methods , Glucan 1,4-alpha-Glucosidase/genetics , Glucan 1,4-alpha-Glucosidase/immunology , Glucan 1,4-alpha-Glucosidase/metabolism , Glycogen Storage Disease Type II/drug therapy , Male , Mice , Mice, Knockout , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
4.
Blood ; 125(19): 2937-47, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25833958

ABSTRACT

CD4(+)CD25(+)FoxP3(+) regulatory T cells (Treg) are critical elements for maintaining immune tolerance, for instance to exogenous antigens that are introduced during therapeutic interventions such as cell/organ transplant or gene/protein replacement therapy. Coadministration of antigen with rapamycin simultaneously promotes deletion of conventional CD4(+) T cells and induction of Treg. Here, we report that the cytokine FMS-like receptor tyrosine kinase ligand (Flt3L) enhances the in vivo effect of rapamycin. This occurs via selective expansion of plasmacytoid dendritic cells (pDCs), which further augments the number of Treg. Whereas in conventional DCs, rapamycin effectively blocks mammalian target of rapamycin (mTOR) 1 signaling induced by Flt3L, increased mTOR1 activity renders pDCs more resistant to inhibition by rapamycin. Consequently, Flt3L and rapamycin synergistically promote induction of antigen-specific Treg via selective expansion of pDCs. This concept is supported by the finding that Treg induction is abrogated upon pDC depletion. The combination with pDCs and rapamycin is requisite for Flt3L/antigen-induced Treg induction because Flt3L/antigen by itself fails to induce Treg. As co-administering Flt3L, rapamycin, and antigen blocked CD8(+) T-cell and antibody responses in models of gene and protein therapy, we conclude that the differential effect of rapamycin on DC subsets can be exploited for improved tolerance induction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Membrane Proteins/metabolism , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Drug Synergism , Flow Cytometry , Humans , Immune Tolerance/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
PLoS One ; 9(6): e98336, 2014.
Article in English | MEDLINE | ID: mdl-24897114

ABSTRACT

Enzyme replacement therapy (ERT) with recombinant human acid-α-glucosidase (rhGAA) is the only FDA approved therapy for Pompe disease. Without ERT, severely affected individuals (early onset) succumb to the disease within 2 years of life. A spectrum of disease severity and progression exists depending upon the type of mutation in the GAA gene (GAA), which in turn determines the amount of defective protein produced and its enzymatic activity. A large percent of the early onset patients are also cross reactive immunological material negative (CRIM-) and develop high titer immune responses to ERT with rhGAA. New insights from our studies in pre-clinical murine models reveal that the type of Gaa mutation has a profound effect on the immune responses mounted against ERT and the associated toxicities, including activation of clotting factors and disseminated intravascular coagulation (DIC). Additionally, the mouse strain affects outcomes, suggesting the influence of additional genetic components or modifiers. High doses of rhGAA (20 mg/kg) are currently required to achieve therapeutic benefit. Our studies indicate that lower enzyme doses reduce the antibody responses to rhGAA, reduce the incidence of immune toxicity and avoid ERT-associated anaphylaxis. Therefore, development of rhGAA with increased efficacy is warranted to limit immunotoxicities.


Subject(s)
Glycogen Storage Disease Type II/immunology , Immunity, Active/genetics , Thrombophilia/genetics , alpha-Glucosidases/therapeutic use , Animals , Cross Reactions/genetics , Dose-Response Relationship, Drug , Enzyme Replacement Therapy , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/genetics , Mice , Mutation
6.
Mol Ther ; 22(4): 702-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24336173

ABSTRACT

Pompe disease is an autosomal recessive disorder caused by mutations in the acid-α glucosidase (GAA) gene. Lingual dysfunction is prominent but does not respond to conventional enzyme replacement therapy (ERT). Using Pompe (Gaa(-/-)) mice, we tested the hypothesis that intralingual delivery of viral vectors encoding GAA results in GAA expression and glycogen clearance in both tongue myofibers and hypoglossal (XII) motoneurons. An intralingual injection of an adeno-associated virus (AAV) vector encoding GAA (serotypes 1 or 9; 1 × 10(11) vector genomes, CMV promoter) was performed in 2-month-old Gaa(-/-) mice, and tissues were harvested 4 months later. Both serotypes robustly transduced tongue myofibers with histological confirmation of GAA expression (immunochemistry) and glycogen clearance (Period acid-Schiff stain). Both vectors also led to medullary transgene expression. GAA-positive motoneurons did not show the histopathologic features which are typical in Pompe disease and animal models. Intralingual injection with the AAV9 vector resulted in approximately threefold more GAA-positive XII motoneurons (P < 0.02 versus AAV1); the AAV9 group also gained more body weight over the course of the study (P < 0.05 versus AAV1 and sham). We conclude that intralingual injection of AAV1 or AAV9 drives persistent GAA expression in tongue myofibers and motoneurons, but AAV9 may more effectively target motoneurons.


Subject(s)
Genetic Therapy , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Motor Neurons/metabolism , alpha-Glucosidases/genetics , Animals , Dependovirus/genetics , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques , Glycogen , Glycogen Storage Disease Type II/pathology , Humans , Injections, Intramuscular , Mice , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Myofibrils/genetics , Myofibrils/metabolism , Promoter Regions, Genetic , alpha-Glucosidases/biosynthesis
7.
EMBO Mol Med ; 5(11): 1698-709, 2013 11.
Article in English | MEDLINE | ID: mdl-24106230

ABSTRACT

Formation of pathogenic antibodies is a major problem in replacement therapies for inherited protein deficiencies. For example, antibodies to coagulation factors ('inhibitors') seriously complicate treatment of haemophilia. While immune tolerance induction (ITI) protocols have been developed, inhibitors against factor IX (FIX) are difficult to eradicate due to anaphylactic reactions and nephrotic syndrome and thus substantially elevate risks for morbidity and mortality. However, hepatic gene transfer with an adeno-associated virus (AAV) serotype 8 vector expressing FIX (at levels of ≥4% of normal) rapidly reversed pre-existing high-titre inhibitors in haemophilia B mice, eliminated antibody production by B cells, desensitized from anaphylaxis (even if protein therapy was resumed) and provided long-term correction. High levels of FIX protein suppressed memory B cells and increased Treg induction, indicating direct and indirect mechanisms of suppression of inhibitor formation. Persistent presence of Treg was required to prevent relapse of antibodies. Together, these data suggest that hepatic gene transfer-based ITI provides a safe and effective alternative to eradicate inhibitors. This strategy may be broadly applicable to reversal of antibodies in different genetic diseases.


Subject(s)
Antibodies/immunology , B-Lymphocytes/immunology , Factor IX/genetics , Genetic Therapy , Hemophilia B/genetics , Hemophilia B/therapy , Animals , Antibody Formation , Dependovirus/genetics , Dependovirus/physiology , Factor IX/immunology , Genetic Vectors/genetics , Genetic Vectors/physiology , Hemophilia B/immunology , Humans , Liver/immunology , Liver/virology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
8.
J Pediatr ; 163(3): 847-54.e1, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23601496

ABSTRACT

OBJECTIVE: To evaluate whether B-cell depletion before enzyme replacement therapy (ERT) initiation can block acid alpha-glucosidase (GAA) antibody responses and improve clinical outcomes. STUDY DESIGN: Six subjects with Pompe disease (including 4 cross-reacting immunologic material-negative infants) aged 2-8 months received rituximab and sirolimus or mycophenolate before ERT. Four subjects continued to receive sirolimus, rituximab every 12 weeks, and intravenous immunoglobulin monthly for the duration of ERT. Sirolimus trough levels, IgG, CD3, CD4, CD8, CD19, CD20, N-terminal pro-brain natriuretic peptide, creatine kinase, creatine kinase-MB, C-reactive protein, platelets, alkaline phosphatase, gamma-glutamyl transferase, aspartate aminotransferase, and alanine aminotransferase were measured regularly. RESULTS: Immunomodulation achieved B-cell depletion without adverse effects. After 17-36 months of rituximab, sirolimus and ERT, all subjects lacked antibodies against GAA, 4 continued to gain motor milestones, yet 2 progressed to require invasive ventilation. The absence of infusion-associated reactions allowed the use of accelerated infusion rates. CONCLUSION: B-cell depletion and T-cell immunomodulation in infants naïve to ERT was accomplished safely and eliminated immune responses against GAA, thereby optimizing clinical outcome; however, this approach did not necessarily influence sustained independent ventilation. Importantly, study outcomes support the initiation of immunomodulation before starting ERT, because the study regimen allowed for prompt initiation of treatment.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Enzyme Replacement Therapy , Glycogen Storage Disease Type II/drug therapy , Immunosuppressive Agents/therapeutic use , Mycophenolic Acid/analogs & derivatives , Sirolimus/therapeutic use , alpha-Glucosidases/therapeutic use , Antigens, CD/blood , Autoantibodies/blood , B-Lymphocytes/metabolism , Biomarkers/blood , Drug Administration Schedule , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/immunology , Humans , Immunoglobulin G/blood , Immunoglobulins, Intravenous/therapeutic use , Infant , Male , Mycophenolic Acid/therapeutic use , Rituximab , Treatment Outcome , alpha-Glucosidases/immunology
9.
Mol Genet Metab ; 106(2): 189-95, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22494547

ABSTRACT

Pompe disease is a neuromuscular disease caused by an inherited deficiency of the lysosomal enzyme acid α-glucosidase (GAA). The resulting accumulation of glycogen causes muscle weakness with the severe form of the disease resulting in death by cardiorespiratory failure in the first year of life. The only available treatment, enzyme replacement therapy (ERT) with recombinant GAA (rhGAA), is severely hampered by antibody responses that reduce efficacy and cause immunotoxicities. Currently, Pompe mice represent the only pre-clinical model for development of new treatments and for immunological studies. While antibody formation following ERT in this model has been described, the underlying T cell response has not been studied. In order to define the T helper response to rhGAA in Pompe mice, immunodominant CD4(+) T cell epitopes were mapped in GAA(-/-) 129SVE mice using ELISpot. Additionally, cytokine responses and antibody formation against rhGAA during ERT were measured. Among the three CD4(+) T cell epitopes identified, only epitope IFLGPEPKSVVQ, predicted to be the strongest MHC II binder, consistently contributed to IL-4 production. Frequencies of IL-4 producing T cells were considerably higher than those of IL-17 or IFN-γ producing cells, suggesting a predominantly Th2 cell mediated response. This is further supported by IgG1 being the prevalent antibody subclass against rhGAA during ERT and consistent with prior reports on IgE formation and anaphylaxis in this model. These results will facilitate mechanistic studies of the immune response to rhGAA in Pompe mice during development of new therapies and tolerance protocols.


Subject(s)
Glycogen Storage Disease Type II/immunology , T-Lymphocytes, Helper-Inducer/immunology , alpha-Glucosidases/immunology , Amino Acid Sequence , Animals , Cytokines/immunology , Disease Models, Animal , Enzyme Replacement Therapy/adverse effects , Epitope Mapping , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Glycogen Storage Disease Type II/therapy , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, 129 Strain , Mice, Knockout , Molecular Sequence Data , Peptide Library , Th2 Cells/immunology , alpha-Glucosidases/therapeutic use
10.
Mol Cell Biol ; 32(10): 1893-907, 2012 May.
Article in English | MEDLINE | ID: mdl-22431524

ABSTRACT

Cancer cells exhibit altered glucose metabolism characterized by a preference for aerobic glycolysis or the Warburg effect, and the cells resist matrix detachment-induced apoptosis, which is called anoikis, a barrier to metastasis. It remains largely unclear whether tumor metabolism influences anoikis and metastasis. Here we show that when detached from the matrix, untransformed mammary epithelial cells undergo metabolic reprogramming by markedly upregulating pyruvate dehydrogenase (PDH) kinase 4 (PDK4) through estrogen-related receptor gamma (ERRγ), thereby inhibiting PDH and attenuating the flux of glycolytic carbon into mitochondrial oxidation. To decipher the significance of this metabolic response, we found that depletion of PDK4 or activation of PDH increased mitochondrial respiration and oxidative stress in suspended cells, resulting in heightened anoikis. Conversely, overexpression of PDKs prolonged survival of cells in suspension. Therefore, decreased glucose oxidation following cell detachment confers anoikis resistance. Unlike untransformed cells, most cancer cells demonstrate reduced glucose oxidation even under attached conditions, and thus they inherently possess a survival advantage when suspended. Normalization of glucose metabolism by stimulating PDH in cancer cells restores their susceptibility to anoikis and impairs their metastatic potential. These results suggest that the Warburg effect, more specifically, diminished glucose oxidation, promotes anoikis resistance and metastasis and that PDKs are potential targets for antimetastasis therapy.


Subject(s)
Anoikis/physiology , Glucose/metabolism , Neoplasm Metastasis , Cell Adhesion/physiology , Cell Line, Tumor , Enzyme Activation , Female , Humans , Mitochondria/metabolism , Oxidation-Reduction , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Receptors, Estrogen/metabolism
11.
Hum Mol Genet ; 20(R1): R61-8, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21518733

ABSTRACT

Pompe disease is an autosomal recessive metabolic myopathy caused by the deficiency of the lysosomal enzyme acid alpha-glucosidase and results in cellular lysosomal and cytoplasmic glycogen accumulation. A wide spectrum of disease exists from hypotonia and severe cardiac hypertrophy in the first few months of life due to severe mutations to a milder form with the onset of symptoms in adulthood. In either condition, the involvement of several systems leads to progressive weakness and disability. In early-onset severe cases, the natural history is characteristically cardiorespiratory failure and death in the first year of life. Since the advent of enzyme replacement therapy (ERT), the clinical outcomes have improved. However, it has become apparent that a new natural history is being defined in which some patients have substantial improvement following ERT, while others develop chronic disability reminiscent of the late-onset disease. In order to improve on the current clinical outcomes in Pompe patients with diminished clinical response to ERT, we sought to address the cause and potential for the treatment of disease manifestations which are not amenable to ERT. In this review, we will focus on the preclinical studies that are relevant to the development of a gene therapy strategy for Pompe disease, and have led to the first clinical trial of recombinant adeno-associated virus-mediated gene-based therapy for Pompe disease. We will cover the preliminary laboratory studies and rationale for a clinical trial, which is based on the treatment of the high rate of respiratory failure in the early-onset patients receiving ERT.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Glycogen Storage Disease Type II/therapy , Clinical Trials as Topic , Enzyme Replacement Therapy , Genetic Vectors/administration & dosage , Glycogen/metabolism , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/pathology , Humans , Treatment Outcome
12.
Front Microbiol ; 2: 244, 2011.
Article in English | MEDLINE | ID: mdl-22279442

ABSTRACT

Intramuscular (IM) administration of an adeno-associated viral (AAV) vector represents a simple and safe method of gene transfer for treatment of the X-linked bleeding disorder hemophilia B (factor IX, F.IX, deficiency). However, the approach is hampered by an increased risk of immune responses against F.IX. Previously, we demonstrated that the drug cocktail of immune suppressants rapamycin, IL-10, and a specific peptide (encoding a dominant CD4(+) T cell epitope) caused an induction of regulatory T cells (Treg) with a concomitant apoptosis of antigen-specific effector T cells (Nayak et al., 2009). This protocol was effective in preventing inhibitory antibody formation against human F.IX (hF.IX) in muscle gene transfer to C3H/HeJ hemophilia B mice (with targeted F9 gene deletion). Here, we show that this protocol can also be used to reverse inhibitor formation. IM injection of AAV1-hF.IX vector resulted in inhibitors of on average 8-10 BU within 1 month. Subsequent treatment with the tolerogenic cocktail accomplished a rapid reduction of hF.IX-specific antibodies to <2 BU, which lasted for >4.5 months. Systemic hF.IX expression increased from undetectable to >200 ng/ml, and coagulation times improved. In addition, we developed an alternative prophylactic protocol against inhibitor formation that did not require knowledge of T cell epitopes, consisting of daily oral administration of rapamycin for 1-month combined with frequent, low-dose intravenous injection of hF.IX protein. Experiments in T cell receptor transgenic mice showed that the route and dosing schedule of drug administration substantially affected Treg induction. When combined with intravenous antigen administration, oral delivery of rapamycin had to be performed daily in order to induce Treg, which were suppressive and phenotypically comparable to natural Treg.

13.
Int Immunol ; 22(4): 259-70, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20139172

ABSTRACT

Naturally occurring regulatory T cells (Treg) express high levels of glucocorticoid-induced tumour necrosis factor receptor (GITR). However, studies of the role of GITR in Treg biology has been complicated by the observation that upon activation effector CD4(+) T (Teff) cells also express the receptor. Here, we dissect the contribution of GITR-induced signaling networks in the expansion and function of FoxP3(+) Treg. We demonstrate that a high-affinity soluble Fc-GITR-L dimer, in conjugation with alphaCD3, specifically enhances in vitro proliferation of Treg, which retain their phenotypic markers (CD25 and FoxP3) and their suppressor function, while minimally affecting Teff cells. Furthermore, Fc-GITR-L does not impair Teff susceptibility to suppression, as judged by cocultures employing GITR-deficient and GITR-sufficient CD4(+) T-cell subsets. Notably, this expansion of Treg could also be seen in vivo, by injecting FoxP3-IRES-GFP mice with Fc-GITR-L even in the absence of antigenic stimulation. In order to test the efficacy of these findings therapeutically, we made use of a C3H/HeJ hemophilia B-prone mouse model. The use of liver-targeted human coagulation factor IX (hF.IX) gene therapy in this model has been shown to induce liver toxicity and the subsequent failure of hF.IX expression. Interestingly, injection of Fc-GITR-L into the hemophilia-prone mice that were undergoing liver-targeted hF.IX gene therapy increased the expression of F.IX and reduced the anticoagulation factors. We conclude that GITR engagement enhances Treg proliferation both in vitro and in vivo and that Fc-GITR-L may be a useful tool for in vivo tolerance induction.


Subject(s)
Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation , Disease Models, Animal , Factor IX/genetics , Forkhead Transcription Factors/metabolism , Genetic Therapy , Glucocorticoid-Induced TNFR-Related Protein , Hemophilia B/therapy , Humans , Immune Tolerance , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Ligands , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Receptors, Nerve Growth Factor/administration & dosage , Receptors, Nerve Growth Factor/genetics , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
PLoS One ; 4(8): e6376, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19652717

ABSTRACT

BACKGROUND: Hepatic gene transfer, in particular using adeno-associated viral (AAV) vectors, has been shown to induce immune tolerance to several protein antigens. This approach has been exploited in animal models of inherited protein deficiency for systemic delivery of therapeutic proteins. Adequate levels of transgene expression in hepatocytes induce a suppressive T cell response, thereby promoting immune tolerance. This study addresses the question of whether AAV gene transfer can induce tolerance to a cytoplasmic protein. MAJOR FINDINGS: AAV-2 vector-mediated hepatic gene transfer for expression of cytoplasmic beta-galactosidase (beta-gal) was performed in immune competent mice, followed by a secondary beta-gal gene transfer with E1/E3-deleted adenoviral Ad-LacZ vector to provoke a severe immunotoxic response. Transgene expression from the AAV-2 vector in approximately 2% of hepatocytes almost completely protected from inflammatory T cell responses against beta-gal, eliminated antibody formation, and significantly reduced adenovirus-induced hepatotoxicity. Consequently, approximately 10% of hepatocytes continued to express beta-gal 45 days after secondary Ad-LacZ gene transfer, a time point when control mice had lost all Ad-LacZ derived expression. Suppression of inflammatory T cell infiltration in the liver and liver damage was linked to specific transgene expression and was not seen for secondary gene transfer with Ad-GFP. A combination of adoptive transfer studies and flow cytometric analyses demonstrated induction of Treg that actively suppressed CD8(+) T cell responses to beta-gal and that was amplified in liver and spleen upon secondary Ad-LacZ gene transfer. CONCLUSIONS: These data demonstrate that tolerance induction by hepatic AAV gene transfer does not require systemic delivery of the transgene product and that expression of a cytoplasmic neo-antigen in few hepatocytes can induce Treg and provide long-term suppression of inflammatory responses and immunotoxicity.


Subject(s)
Cytoplasm/enzymology , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Immune Tolerance/genetics , Liver/metabolism , beta-Galactosidase/immunology , Animals , Liver/enzymology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transduction, Genetic , beta-Galactosidase/genetics
15.
Mol Ther ; 17(10): 1733-42, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19603001

ABSTRACT

Immune responses to factor IX (F.IX), a major concern in gene therapy for hemophilia, were analyzed for adeno-associated viral (AAV-2) gene transfer to skeletal muscle and liver as a function of the F9 underlying mutation. Vectors identical to those recently used in clinical trials were administered to four lines of hemophilia B mice on a defined genetic background [C3H/HeJ with deletion of endogenous F9 and transgenic for a range of nonfunctional human F.IX (hF.IX) variants]. The strength of the immune response to AAV-encoded F.IX inversely correlated with the degree of conservation of endogenous coding information and levels of endogenous antigen. Null mutation animals developed T- and B-cell responses in both protocols. However, inhibitor titers were considerably higher upon muscle gene transfer (or protein therapy). Transduced muscles of Null mice had strong infiltrates with CD8+ cells, which were much more limited in the liver and not seen for the other mutations. Sustained expression was achieved with liver transduction in mice with crm(-) nonsense and missense mutations, although they still formed antibodies upon muscle gene transfer. Therefore, endogenous expression prevented T-cell responses more effectively than antibody formation, and immune responses varied substantially depending on the protocol and the underlying mutation.


Subject(s)
Factor IX/immunology , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hemophilia B/therapy , Adenoviridae/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Factor IX/genetics , Genetic Therapy/adverse effects , Humans , Immunity, Humoral/genetics , Immunity, Humoral/immunology , Immunohistochemistry , Mice , Microscopy, Fluorescence , Mutation , Mutation, Missense
16.
Hum Gene Ther ; 20(7): 767-76, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19309290

ABSTRACT

Gene therapy for hemophilia B has been shown to result in long-term expression and immune tolerance to factor IX (F.IX) after in vivo transduction of hepatocytes with adeno-associated viral (AAV-2) vectors in experimental animals. An optimized protocol was effective in several strains of mice with a factor 9 gene deletion (F9(-/-)). However, immune responses against F.IX were repeatedly observed in C3H/HeJ F9(-/-) mice. We sought to establish a gene transfer protocol that results in sustained expression without a requirement for additional manipulation of the immune system. Compared with AAV-2, AAV-8 was more efficient in transgene expression and induction of tolerance to F.IX in three different strains of wild-type mice. At equal vector doses, AAV-8 induced transgene product-specific regulatory CD4(+)CD25(+)FoxP3(+) T cells at significantly higher frequency. Moreover, sustained correction of hemophilia B in C3H/HeJ F9(-/-) mice without antibody formation was documented in all animals treated with > or =4 x 10(11) vector genomes (VG)/kg and in 80% of mice treated with 8 x 10(10) VG/kg. Therefore, it is possible to develop a gene transfer protocol that reliably induces tolerance to F.IX largely independent of genetic factors. A comparison with other studies suggests that additional parameters besides plateau levels of F.IX expression contributed to the improved success rate of tolerance induction.


Subject(s)
Dependovirus/genetics , Factor IX/genetics , Factor IX/immunology , Gene Transfer Techniques , Genetic Therapy , Immune Tolerance/immunology , Liver/metabolism , Animals , Antibody Formation/immunology , Factor IX/therapeutic use , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hemophilia B/genetics , Hemophilia B/immunology , Hemophilia B/therapy , Humans , Immunoglobulin G/biosynthesis , Liver/pathology , Mice , T-Lymphocytes, Regulatory/immunology , Time Factors , Transgenes/genetics
17.
Blood ; 110(4): 1132-40, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17438084

ABSTRACT

Gene replacement therapy is complicated by the risk of an immune response against the therapeutic transgene product, which in part is determined by the route of vector administration. Our previous studies demonstrated induction of immune tolerance to coagulation factor IX (FIX) by hepatic adeno-associated viral (AAV) gene transfer. Using a regulatory T-cell (T(reg))-deficient model (Rag-2(-/-) mice transgenic for ovalbumin-specific T-cell receptor DO11.10), we provide first definitive evidence for induction of transgene product-specific CD4(+)CD25(+) T(regs) by in vivo gene transfer. Hepatic gene transfer-induced T(regs) express FoxP3, GITR, and CTLA4, and suppress CD4(+)CD25(-) T cells. T(regs) are detected as early as 2 weeks after gene transfer, and increase in frequency in thymus and secondary lymphoid organs during the following 2 months. Similarly, adoptive lymphocyte transfers from mice tolerized to human FIX by hepatic AAV gene transfer indicate induction of CD4(+)CD25(+)GITR(+) that suppresses antibody formation to FIX. Moreover, in vivo depletion of CD4(+)CD25(+) T(regs) leads to antibody formation to the FIX transgene product after hepatic gene transfer, which strongly suggests that these regulatory cells are required for tolerance induction. Our study reveals a crucial role of CD4(+)CD25(+) T(regs) in preventing immune responses to the transgene product in gene transfer.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/physiology , Factor IX/administration & dosage , Gene Transfer Techniques , Interleukin-2 Receptor alpha Subunit/immunology , Liver/immunology , T-Lymphocytes, Regulatory/immunology , Animals , DNA-Binding Proteins/genetics , Dependovirus/genetics , Factor IX/immunology , Factor IX/metabolism , Forkhead Transcription Factors , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Glucocorticoid-Induced TNFR-Related Protein , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Humans , Immune Tolerance , Immunization , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptors, Nerve Growth Factor , Receptors, Tumor Necrosis Factor , Spleen/cytology , Spleen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...