Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Allergy Clin Immunol ; 140(6): 1660-1670.e16, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28400115

ABSTRACT

BACKGROUND: Thymus transplantation is a promising strategy for the treatment of athymic complete DiGeorge syndrome (cDGS). METHODS: Twelve patients with cDGS underwent transplantation with allogeneic cultured thymus. OBJECTIVE: We sought to confirm and extend the results previously obtained in a single center. RESULTS: Two patients died of pre-existing viral infections without having thymopoiesis, and 1 late death occurred from autoimmune thrombocytopenia. One infant had septic shock shortly after transplantation, resulting in graft loss and the need for a second transplant. Evidence of thymopoiesis developed from 5 to 6 months after transplantation in 10 patients. Median circulating naive CD4 counts were 44 × 106/L (range, 11-440 × 106/L) and 200 × 106/L (range, 5-310 × 106/L) at 12 and 24 months after transplantation and T-cell receptor excision circles were 2,238/106 T cells (range, 320-8,807/106 T cells) and 4,184/106 T cells (range, 1,582-24,596/106 T cells). Counts did not usually reach normal levels for age, but patients were able to clear pre-existing infections and those acquired later. At a median of 49 months (range, 22-80 months), 8 have ceased prophylactic antimicrobials, and 5 have ceased immunoglobulin replacement. Histologic confirmation of thymopoiesis was seen in 7 of 11 patients undergoing biopsy of transplanted tissue, including 5 showing full maturation through to the terminal stage of Hassall body formation. Autoimmune regulator expression was also demonstrated. Autoimmune complications were seen in 7 of 12 patients. In 2 patients early transient autoimmune hemolysis settled after treatment and did not recur. The other 5 experienced ongoing autoimmune problems, including thyroiditis (3), hemolysis (1), thrombocytopenia (4), and neutropenia (1). CONCLUSIONS: This study confirms the previous reports that thymus transplantation can reconstitute T cells in patients with cDGS but with frequent autoimmune complications in survivors.


Subject(s)
Autoimmune Diseases/immunology , DiGeorge Syndrome/therapy , Organ Transplantation , Postoperative Complications/immunology , T-Lymphocytes/immunology , Thymus Gland/transplantation , Autoimmune Diseases/etiology , Cells, Cultured , Child , Child, Preschool , DiGeorge Syndrome/immunology , Europe , Female , Humans , Immune Reconstitution , Infant , Male , Organ Culture Techniques , Transplantation, Homologous , Treatment Outcome
2.
Blood ; 118(9): 2492-501, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21690559

ABSTRACT

Rearrangement of the cytoskeleton in T cells plays a critical role in the organization of a complex signaling interface referred to as immunologic synapse (IS). Surprisingly, the contribution of antigen presenting cells, in particular dendritic cells (DCs), to the structure and function of the IS has not been investigated in as much detail. We have used a natural model of cytoskeletal dysfunction caused by deficiency of the Wiskott-Aldrich syndrome protein (WASp) to explore the contribution of the DC cytoskeleton to IS formation and to T-cell priming. In an antigen-specific system, T-DC contacts were found to be less stable when DCs alone lacked WASp, and associated with multiple defects of IS structure. As a consequence, DCs were unable to support normal IL-12 secretion, and events downstream of TCR signaling were abrogated, including increased calcium flux, microtubule organizing center (MTOC) polarization, phosphorylation of ZAP-70, and T-cell proliferation. Formation of an effective signaling interface is therefore dependent on active cytoskeletal rearrangements in DCs even when T cells are functionally competent. Deficiency of DC-mediated activities may contribute significantly to the varied immunodysregulation observed in patients with WAS, and also in those with limited myeloid reconstitution after allogeneic hematopoietic stem cell transplantation.


Subject(s)
Cytoskeleton/ultrastructure , Dendritic Cells/ultrastructure , Immunological Synapses/ultrastructure , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein/physiology , Animals , Bacterial Proteins/analysis , Bacterial Proteins/genetics , Calcium Signaling/immunology , Cell Movement , Crosses, Genetic , Genes, Reporter , Genetic Complementation Test , Humans , Immunological Synapses/immunology , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , T-Lymphocytes/ultrastructure , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/immunology
3.
Mol Ther ; 19(1): 122-32, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20978475

ABSTRACT

X-linked chronic granulomatous disease (X-CGD) is a primary immunodeficiency caused by mutations in the CYBB gene encoding the phagocyte nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase catalytic subunit gp91(phox). A recent clinical trial for X-CGD using a spleen focus-forming virus (SFFV)-based γ-retroviral vector has demonstrated clear therapeutic benefits in several patients although complicated by enhancer-mediated mutagenesis and diminution of effectiveness over time due to silencing of the viral long terminal repeat (LTR). To improve safety and efficacy, we have designed a lentiviral vector that directs transgene expression primarily in myeloid cells. To this end, we created a synthetic chimeric promoter that contains binding sites for myeloid transcription factors CAAT box enhancer-binding family proteins (C/EBPs) and PU.1, which are highly expressed during granulocytic differentiation. As predicted, the chimeric promoter regulated higher reporter gene expression in myeloid than in nonmyeloid cells, and in human hematopoietic progenitors upon granulocytic differentiation. In a murine model of stem cell gene therapy for X-CGD, the chimeric vector resulted in high levels of gp91(phox) expression in committed myeloid cells and granulocytes, and restored normal NADPH-oxidase activity. These findings were recapitulated in human neutrophils derived from transduced X-CGD CD34(+) cells in vivo, and suggest that the chimeric promoter will have utility for gene therapy of myeloid lineage disorders such as CGD.


Subject(s)
Cathepsin G/genetics , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/therapy , Myeloid Cells/physiology , Proto-Oncogene Proteins c-fes/genetics , Recombinant Fusion Proteins/genetics , Transgenes , Animals , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , DNA Copy Number Variations , Enhancer Elements, Genetic , Gene Expression Regulation , Genes, X-Linked , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/metabolism , Granulocytes/metabolism , Granulomatous Disease, Chronic/enzymology , Granulomatous Disease, Chronic/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , Molecular Sequence Data , Mutagenesis/genetics , Myeloid Cells/metabolism , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Retroviridae/metabolism , Spleen Focus-Forming Viruses/genetics , Spleen Focus-Forming Viruses/metabolism , Stem Cells/metabolism , Terminal Repeat Sequences , Trans-Activators/metabolism
4.
J Clin Invest ; 118(9): 3143-50, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18688286

ABSTRACT

X-linked SCID (SCID-X1) is amenable to correction by gene therapy using conventional gammaretroviral vectors. Here, we describe the occurrence of clonal T cell acute lymphoblastic leukemia (T-ALL) promoted by insertional mutagenesis in a completed gene therapy trial of 10 SCID-X1 patients. Integration of the vector in an antisense orientation 35 kb upstream of the protooncogene LIM domain only 2 (LMO2) caused overexpression of LMO2 in the leukemic clone. However, leukemogenesis was likely precipitated by the acquisition of other genetic abnormalities unrelated to vector insertion, including a gain-of-function mutation in NOTCH1, deletion of the tumor suppressor gene locus cyclin-dependent kinase 2A (CDKN2A), and translocation of the TCR-beta region to the STIL-TAL1 locus. These findings highlight a general toxicity of endogenous gammaretroviral enhancer elements and also identify a combinatorial process during leukemic evolution that will be important for risk stratification and for future protocol design.


Subject(s)
Chromosomes, Human, X , Genetic Therapy/adverse effects , Genetic Therapy/methods , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Severe Combined Immunodeficiency/therapy , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA-Binding Proteins/genetics , Follow-Up Studies , Humans , Infant , LIM Domain Proteins , Male , Metalloproteins/genetics , Models, Biological , Mutagenesis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Proto-Oncogene Proteins , Receptor, Notch1/genetics , Receptors, Interleukin-2/genetics , Severe Combined Immunodeficiency/complications
5.
J Clin Invest ; 117(8): 2241-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17671654

ABSTRACT

We treated 10 children with X-linked SCID (SCID-X1) using gammaretrovirus-mediated gene transfer. Those with sufficient follow-up were found to have recovered substantial immunity in the absence of any serious adverse events up to 5 years after treatment. To determine the influence of vector integration on lymphoid reconstitution, we compared retroviral integration sites (RISs) from peripheral blood CD3(+) T lymphocytes of 5 patients taken between 9 and 30 months after transplantation with transduced CD34(+) progenitor cells derived from 1 further patient and 1 healthy donor. Integration occurred preferentially in gene regions on either side of transcription start sites, was clustered, and correlated with the expression level in CD34(+) progenitors during transduction. In contrast to those in CD34(+) cells, RISs recovered from engrafted CD3(+) T cells were significantly overrepresented within or near genes encoding proteins with kinase or transferase activity or involved in phosphorus metabolism. Although gross patterns of gene expression were unchanged in transduced cells, the divergence of RIS target frequency between transduced progenitor cells and post-thymic T lymphocytes indicates that vector integration influences cell survival, engraftment, or proliferation.


Subject(s)
CD3 Complex , Gammaretrovirus , Genetic Vectors , Hematopoietic Stem Cell Transplantation , T-Lymphocytes/immunology , Virus Integration , X-Linked Combined Immunodeficiency Diseases/therapy , Adult , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Child , Child, Preschool , Female , Follow-Up Studies , Graft Survival/genetics , Graft Survival/immunology , Hematopoietic Stem Cells/immunology , Humans , Infant , Male , Transduction, Genetic , Transplantation, Autologous , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
6.
Lancet ; 364(9452): 2181-7, 2004.
Article in English | MEDLINE | ID: mdl-15610804

ABSTRACT

BACKGROUND: X-linked severe combined immunodeficiency (SCID-X1) is caused by mutations in the common cytokine-receptor gamma chain (gamma(c)), resulting in disruption of development of T lymphocytes and natural-killer cells. B-lymphocyte function is also intrinsically compromised. Allogeneic bone-marrow transplantation is successful if HLA-matched family donors are available, but HLA-mismatched procedures are associated with substantial morbidity and mortality. We investigated the application of somatic gene therapy by use of a gibbon-ape-leukaemia-virus pseudotyped gammaretroviral vector. METHODS: Four children with SCID-X1 were enrolled. Autologous CD34-positive haemopoietic bone-marrow stem cells were transduced ex vivo and returned to the patients without preceding cytoreductive chemotherapy. The patients were monitored for integration and expression of the gamma(c) vector and for functional immunological recovery. FINDINGS: All patients have shown substantial improvements in clinical and immunological features, and prophylactic medication could be withdrawn in two. No serious adverse events have been recorded. T cells responded normally to mitogenic and antigenic stimuli, and the T-cell-receptor (TCR) repertoire was highly diverse. Where assessable, humoral immunity, in terms of antibody production, was also restored and associated with increasing rates of somatic mutation in immunoglobulin genes. INTERPRETATION: Gene therapy for SCID-X1 is a highly effective strategy for restoration of functional cellular and humoral immunity.


Subject(s)
Genetic Diseases, X-Linked/therapy , Genetic Therapy , Severe Combined Immunodeficiency/therapy , Antigens, CD34/analysis , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Child, Preschool , Gammaretrovirus , Gene Transfer Techniques , Genetic Diseases, X-Linked/genetics , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors , Humans , Immunity , Immunoglobulins/blood , Infant , Interleukin Receptor Common gamma Subunit , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mutation , Receptors, Interleukin-7/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology , Transduction, Genetic
7.
Sci Transl Med ; 3(97): 97ra79, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865537

ABSTRACT

X-linked severe combined immunodeficiency (SCID-X1) is caused by mutations in the common cytokine receptor γ chain. These mutations classically lead to complete absence of functional T and natural killer cell lineages as well as to intrinsically compromised B cell function. Although human leukocyte antigen (HLA)-matched hematopoietic stem cell transplantation (HSCT) is highly successful in SCID-X1 patients, HLA-mismatched procedures can be associated with prolonged immunodeficiency, graft-versus-host disease, and increased overall mortality. Here, 10 children were treated with autologous CD34(+) hematopoietic stem and progenitor cells transduced with a conventional gammaretroviral vector. The patients did not receive myelosuppressive conditioning and were monitored for immunological recovery after cell infusion. All patients were alive after a median follow-up of 80 months (range, 54 to 107 months), and a functional polyclonal T cell repertoire was restored in all patients. Humoral immunity only partially recovered but was sufficient in some patients to allow for withdrawal of immunoglobulin replacement; however, three patients developed antibiotic-responsive acute pulmonary infection after discontinuation of antibiotic prophylaxis and/or immunoglobulin replacement. One patient developed acute T cell acute lymphoblastic leukemia because of up-regulated expression of the proto-oncogene LMO-2 from insertional mutagenesis, but maintained a polyclonal T cell repertoire through chemotherapy and entered remission. Therefore, gene therapy for SCID-X1 without myelosuppressive conditioning effectively restored T cell immunity and was associated with high survival rates for up to 9 years. Further studies using vectors designed to limit mutagenesis and strategies to enhance B cell reconstitution are warranted to define the role of this treatment modality alongside conventional HSCT for SCID-X1.


Subject(s)
Genetic Therapy/methods , T-Lymphocytes/immunology , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/therapy , Antigens, CD34/genetics , Antigens, CD34/metabolism , Child, Preschool , Female , Gammaretrovirus/genetics , Genetic Vectors/genetics , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Male , Proto-Oncogene Mas , Stem Cell Transplantation/methods , Transplantation, Autologous/methods , X-Linked Combined Immunodeficiency Diseases/metabolism
8.
Sci Transl Med ; 3(97): 97ra80, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865538

ABSTRACT

Genetic defects in the purine salvage enzyme adenosine deaminase (ADA) lead to severe combined immunodeficiency (SCID) with profound depletion of T, B, and natural killer cell lineages. Human leukocyte antigen-matched allogeneic hematopoietic stem cell transplantation (HSCT) offers a successful treatment option. However, individuals who lack a matched donor must receive mismatched transplants, which are associated with considerable morbidity and mortality. Enzyme replacement therapy (ERT) for ADA-SCID is available, but the associated suboptimal correction of immunological defects leaves patients susceptible to infection. Here, six children were treated with autologous CD34-positive hematopoietic bone marrow stem and progenitor cells transduced with a conventional gammaretroviral vector encoding the human ADA gene. All patients stopped ERT and received mild chemotherapy before infusion of gene-modified cells. All patients survived, with a median follow-up of 43 months (range, 24 to 84 months). Four of the six patients recovered immune function as a result of engraftment of gene-corrected cells. In two patients, treatment failed because of disease-specific and technical reasons: Both restarted ERT and remain well. Of the four reconstituted patients, three remained off enzyme replacement. Moreover, three of these four patients discontinued immunoglobulin replacement, and all showed effective metabolic detoxification. All patients remained free of infection, and two cleared problematic persistent cytomegalovirus infection. There were no adverse leukemic side effects. Thus, gene therapy for ADA-SCID is safe, with effective immunological and metabolic correction, and may offer a viable alternative to conventional unrelated donor HSCT.


Subject(s)
Adenosine Deaminase/metabolism , Agammaglobulinemia/therapy , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology , Agammaglobulinemia/immunology , Child, Preschool , Female , Gene Dosage/genetics , Genetic Vectors/genetics , Humans , Infant , Male , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology
9.
Mol Ther ; 12(3): 537-46, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16099415

ABSTRACT

The use of lentiviral vectors for gene transfer into hematopoietic stem cells has raised considerable interest as these vectors can permanently integrate their genome into quiescent cells. Vectors based on alternative lentiviruses would theoretically be safer than HIV-1-based vectors and could also be used in HIV-positive patients, minimizing the risk of generating replication-competent virus. Here we report the use of third-generation equine infectious anemia virus (EIAV)- and HIV-1-based vectors with minimal viral sequences and absence of accessory proteins. We have compared their efficiency in transducing mouse and human hematopoietic stem cells both in vitro and in vivo to that of a previously documented second-generation HIV-1 vector. The third-generation EIAV- and HIV-based vectors gave comparable levels of transduction and transgene expression in both mouse and human NOD/SCID repopulating cells but were less efficient than the second-generation HIV-1 vector in human HSCs. For the EIAV vector this is possibly a reflection of the lower protein expression levels achieved in human cells, as vector copy number analysis revealed that this vector exhibited a trend to integrate equally efficiently compared to the third-generation HIV-1 vector in both mouse and human HSCs. Interestingly, the presence or absence of Tat in viral preparations did not influence the transduction efficiency of HIV-1 vectors in human HSCs.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/instrumentation , Genetic Vectors , HIV/genetics , Hematopoietic Stem Cells/metabolism , Infectious Anemia Virus, Equine/genetics , Animals , Antigens, CD34/biosynthesis , Cell Line , Flow Cytometry , Genetic Therapy/methods , Green Fluorescent Proteins/metabolism , Humans , In Vitro Techniques , Lentivirus/genetics , Lentivirus/metabolism , Leukocyte Common Antigens/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL