Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
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
2.
EMBO Mol Med ; 9(11): 1558-1573, 2017 11.
Article in English | MEDLINE | ID: mdl-28835507

ABSTRACT

Lentiviral vectors (LV) are powerful and versatile vehicles for gene therapy. However, their complex biological composition challenges large-scale manufacturing and raises concerns for in vivo applications, because particle components and contaminants may trigger immune responses. Here, we show that producer cell-derived polymorphic class-I major histocompatibility complexes (MHC-I) are incorporated into the LV surface and trigger allogeneic T-cell responses. By disrupting the beta-2 microglobulin gene in producer cells, we obtained MHC-free LV with substantially reduced immunogenicity. We introduce this targeted editing into a novel stable LV packaging cell line, carrying single-copy inducible vector components, which can be reproducibly converted into high-yield LV producers upon site-specific integration of the LV genome of interest. These LV efficiently transfer genes into relevant targets and are more resistant to complement-mediated inactivation, because of reduced content of the vesicular stomatitis virus envelope glycoprotein G compared to vectors produced by transient transfection. Altogether, these advances support scalable manufacturing of alloantigen-free LV with higher purity and increased complement resistance that are better suited for in vivo gene therapy.


Subject(s)
Gene Editing/methods , Genetic Vectors/metabolism , Lentivirus/genetics , Animals , CD55 Antigens/metabolism , Cell Line , Factor IX/genetics , Factor IX/metabolism , Genetic Therapy , Genetic Vectors/genetics , HEK293 Cells , Hemophilia B/therapy , Humans , Isoantigens/immunology , Membrane Cofactor Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Complement 3b/metabolism , Transfection
3.
EMBO Mol Med ; 9(9): 1198-1211, 2017 09.
Article in English | MEDLINE | ID: mdl-28667090

ABSTRACT

Clinical application of lentiviral vector (LV)-based hematopoietic stem and progenitor cells (HSPC) gene therapy is rapidly becoming a reality. Nevertheless, LV-mediated signaling and its potential functional consequences on HSPC biology remain poorly understood. We unravel here a remarkably limited impact of LV on the HSPC transcriptional landscape. LV escaped innate immune sensing that instead led to robust IFN responses upon transduction with a gamma-retroviral vector. However, reverse-transcribed LV DNA did trigger p53 signaling, activated also by non-integrating Adeno-associated vector, ultimately leading to lower cell recovery ex vivo and engraftment in vivo These effects were more pronounced in the short-term repopulating cells while long-term HSC frequencies remained unaffected. Blocking LV-induced signaling partially rescued both apoptosis and engraftment, highlighting a novel strategy to further dampen the impact of ex vivo gene transfer on HSPC. Overall, our results shed light on viral vector sensing in HSPC and provide critical insight for the development of more stealth gene therapy strategies.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Hematopoietic Stem Cells/immunology , Lentivirus/genetics , Tumor Suppressor Protein p53/immunology , Animals , Genetic Vectors/immunology , Hematopoietic Stem Cell Transplantation , Humans , Immunity, Innate , Lentivirus/immunology , Mice , Tumor Suppressor Protein p53/genetics
4.
Mol Ther ; 23(2): 352-62, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25270076

ABSTRACT

Improving hematopoietic stem and progenitor cell (HSPC) permissiveness to HIV-derived lentiviral vectors (LVs) remains a challenge for the field of gene therapy as high vector doses and prolonged ex vivo culture are still required to achieve clinically relevant transduction levels. We report here that Cyclosporin A (CsA) and Rapamycin (Rapa) significantly improve LV gene transfer in human and murine HSPC. Both compounds increased LV but not gammaretroviral transduction and acted independently of calcineurin and autophagy. Improved gene transfer was achieved across all CD34(+) subpopulations, including in long-term SCID repopulating cells. Effects of CsA were specific of HSPC and opposite to its known impact on HIV replication. Mutating the Cyclophilin A binding pocket of the viral capsid (CA) further improved transduction in combination with CsA. Tracking of the LV genome fate revealed that CsA relieves a CA-dependent early block and increases integration, while Rapa acts early in LV infection independently of the viral CA. In agreement, only Rapa was able to improve transduction by an integrase-defective LV harboring wild-type CA. Overall, our findings pave the way for more efficient and sustainable LV gene therapy in human HSPCs and shed light on the multiple innate barriers specifically hampering LV transduction in these cells.


Subject(s)
Cyclosporine/pharmacology , Genetic Vectors/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Sirolimus/pharmacology , Transduction, Genetic , Animals , Cell Differentiation , Fetal Blood/cytology , Gene Expression , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Immunophenotyping , Mice , Phenotype , Transgenes
5.
EMBO Mol Med ; 5(11): 1684-97, 2013 11.
Article in English | MEDLINE | ID: mdl-24106222

ABSTRACT

A major complication of factor replacement therapy for haemophilia is the development of anti-factor neutralizing antibodies (inhibitors). Here we show that liver gene therapy by lentiviral vectors (LVs) expressing factor IX (FIX) strongly reduces pre-existing anti-FIX antibodies and eradicates FIX inhibitors in haemophilia B mice. Concomitantly, plasma FIX levels and clotting activity rose to 50-100% of normal. The treatment was effective in 75% of treated mice. FIX-specific plasma cells (PCs) and memory B cells were reduced, likely because of memory B-cell depletion in response to constant exposure to high doses of FIX. Regulatory T cells displaying FIX-specific suppressive capacity were induced in gene therapy treated mice and controlled FIX-specific T helper cells. Gene therapy proved safer than a regimen mimicking immune tolerance induction (ITI) by repeated high-dose FIX protein administration, which induced severe anaphylactoid reactions in inhibitors-positive haemophilia B mice. Liver gene therapy can thus reverse pre-existing immunity, induce active tolerance to FIX and establish sustained FIX activity at therapeutic levels. These data position gene therapy as an attractive treatment option for inhibitors-positive haemophilic patients.


Subject(s)
Factor IX/genetics , Genetic Therapy , Hemophilia B/genetics , Hemophilia B/therapy , Lentivirus/physiology , Liver/virology , Animals , Antibodies/immunology , B-Lymphocytes/immunology , Cell Line , Factor IX/immunology , Genetic Vectors/genetics , Genetic Vectors/physiology , Hemophilia B/immunology , Humans , Lentivirus/genetics , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...