scholarly journals 1134. Towards Gene Therapy in Reproduction: In Vitro and In Vivo Gene Transfer to Human and Murine Endometrium Using Adenoviral Vectors

2002 ◽  
Vol 5 (5) ◽  
pp. S368
2003 ◽  
Vol 8 (5) ◽  
pp. 813-821 ◽  
Author(s):  
Fuminori Sakurai ◽  
Hiroyuki Mizuguchi ◽  
Teruhide Yamaguchi ◽  
Takao Hayakawa

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3249-3249
Author(s):  
Barbara Cassani ◽  
Grazia Andolfi ◽  
Massimiliano Mirolo ◽  
Luca Biasco ◽  
Alessandra Recchia ◽  
...  

Abstract Gene transfer into hematopoietic stem/progenitor cells (HSC) by gammaretroviral vectors is an effective treatment for patients affected by severe combined immunodeficiency (SCID) due to adenosine deaminase (ADA)-deficiency. Recent studied have indicated that gammaretroviral vectors integrate in a non-random fashion in their host genome, but there is still limited information on the distribution of retroviral insertion sites (RIS) in human long-term reconstituting HSC following therapeutic gene transfer. We performed a genome-wide analysis of RIS in transduced bone marrow-derived CD34+ cells before transplantation (in vitro) and in hematopoietic cell subsets (ex vivo) from five ADA-SCID patients treated with gene therapy combined to low-dose busulfan. Vector-genome junctions were cloned by inverse or linker-mediated PCR, sequenced, mapped onto the human genome, and compared to a library of randomly cloned human genome fragments or to the expected distribution for the NCBI annotation. Both in vitro (n=212) and ex vivo (n=496) RIS showed a non-random distribution, with strong preference for a 5-kb window around transcription start sites (23.6% and 28.8%, respectively) and for gene-dense regions. Integrations occurring inside the transcribed portion of a RefSeq genes were more represented in vitro than ex vivo (50.9 vs 41.3%), while RIS <30kb upstream from the start site were more frequent in the ex vivo sample (25.6% vs 19.4%). Among recurrently hit loci (n=50), LMO2 was the most represented, with one integration cloned from pre-infusion CD34+ cells and five from post-gene therapy samples (2 in granulocytes, 3 in T cells). Clone-specific Q-PCR showed no in vivo expansion of LMO2-carrying clones while LMO2 gene overexpression at the bulk level was excluded by RT-PCR. Gene expression profiling revealed a preference for integration into genes transcriptionally active in CD34+ cells at the time of transduction as well as genes expressed in T cells. Functional clustering analysis of genes hit by retroviral vectors in pre- and post-transplant cells showed no in vivo skewing towards genes controlling self-renewal or survival of HSC (i.e. cell cycle, transcription, signal transduction). Clonal analysis of long-term repopulating cells (>=6 months) revealed a high number of distinct RIS (range 42–121) in the T-cell compartment, in agreement with the complexity of the T-cell repertoire, while fewer RIS were retrieved from granulocytes. The presence of shared integrants among multiple lineages confirmed that the gene transfer protocol was adequate to allow stable engraftment of multipotent HSC. Taken together, our data show that transplantation of ADA-transduced HSC does not result in skewing or expansion of malignant clones in vivo, despite the occurrence of insertions near potentially oncogenic genomic sites. These results, combined to the relatively long-term follow-up of patients, indicate that retroviral-mediated gene transfer for ADA-SCID has a favorable safety profile.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 197-197
Author(s):  
Masami Niiya ◽  
Masayuki Endo ◽  
Philip W. Zoltick ◽  
Nidal E. Muvarak ◽  
David G. Motto ◽  
...  

Abstract ADAMTS13, a member of A Disintegrin and Metalloprotease with ThromboSpondin type 1 repeats (ADAMTS) family, is mainly synthesized in the hepatic stellate cells, endothelial cells and megakaryocytes or platelets. It controls the sizes of von Willebrand factor (VWF) multimers by cleaving VWF at the Tyr1605-Met1606 bond. Genetic deficiency of plasma ADAMTS13 activity results in hereditary thrombotic thrombocytopenic purpura (TTP), also named Upshaw-Schülman syndrome. To develop a potential gene therapy approach and to determine the domains of ADAMTS13 required for recognition and cleavage of VWF in vivo, a self-inactivating lentiviral vector encoding human wild-type ADAMTS13 or variant truncated after the spacer domain (construct MDTCS) was administrated by intra-amniotic injection on embryonic day 8. Direct stereomicroscopy and immunofluorescent microscopic analysis revealed that the green fluorescent protein (GFP) reporter, ADAMTS13 and MDTCS were predominantly expressed in the heart, kidneys and skin. The synthesized ADAMTS13 and truncated variant were detectable in mouse plasma by immunoprecipitation and Western blot, as well as by proteolytic cleavage of FRETS-VWF73 substrate. The levels of proteolytic activity in plasma of mice expressing ADAMTS13 and MDTCS were 5 ± 7% and 60 ± 70%, respectively using normal human plasma as a standard, and this proteolytic activity persisted for at least 24 weeks in Adamts13−/−mice and 42 weeks in wild-type mice tested (the duration of observation). The mice expressing both recombinant ADAMTS13 and MDTCS showed a significantly decreased ratio of plasma VWF collagen-binding activity to antigen and a reduction in VWF multimer sizes as compared to those in the controls. Moreover, the mice expressing ADAMTS13 and MDTCS showed a significant prolongation of ferric chloride-induced carotid arterial occlusion time (9.0 ± 0.6 and 25.2 ± 3.2 min, respectively) as compared to the Adamts13−/− mice expressing GFP alone (5.6 ± 0.5 min) (p&lt;0.01). The ferric chloride-induced carotid occlusion time in Adamts13−/− mice expressing ADAMTS13 was almost identical to that in wild type mice with same genetic background (C56BL/6) (8.0 ± 0.2 min) (p&gt;0.05). The data demonstrate the correction of the prothrombotic phenotype in Adamts13−/−mice by gene transfer to the fetus by viral vectors encoding human wild type ADAMTS13 and the carboxyl terminal truncated variant (MDTCS), supporting the feasibility of developing a gene therapy based treatment for hereditary TTP. The discrepancy in the proteolytic activity of MDTCS between in vitro (Zhang P et al. Blood, 2007 in press) and in vivo in the present study suggests the potential cofactors in murine circulation that may rescue the defective proteolytic activity of the carboxyl-terminal truncated ADAMTS13 protease seen in vitro.


Blood ◽  
2003 ◽  
Vol 101 (10) ◽  
pp. 3924-3932 ◽  
Author(s):  
Lingfei Xu ◽  
Cuihua Gao ◽  
Mark S. Sands ◽  
Shi-Rong Cai ◽  
Timothy C. Nichols ◽  
...  

AbstractHemophilia B is a bleeding disorder resulting from factor IX (FIX) deficiency that might be treated with gene therapy. Neonatal delivery would correct the disease sooner than would transfer into adults, and could reduce immunological responses. Neonatal mice were injected intravenously with a Moloney murine leukemia virus–based retroviral vector (RV) expressing canine FIX (cFIX). They achieved 150% to 280% of normal cFIX antigen levels in plasma (100% is 5 μg/mL), which was functional in vitro and in vivo. Three newborn hemophilia B dogs that were injected intravenously with RV achieved 12% to 36% of normal cFIX antigen levels, which improved coagulation tests. Only one mild bleed has occurred during 14 total months of evaluation. This is the first demonstration of prolonged expression after neonatal gene therapy for hemophilia B in mice or dogs. Most animals failed to make antibodies to cFIX, demonstrating that neonatal gene transfer may induce tolerance. Although hepatocytes from newborns replicate, those from adults do not. Adult mice therefore received hepatocyte growth factor to induce hepatocyte replication prior to intravenous injection of RV. This resulted in expression of 35% of normal cFIX antigen levels for 11 months, although all mice produced anti-cFIX antibodies. This is the first demonstration that high levels of FIX activity can be achieved with an RV in adults without a partial hepatectomy to induce hepatocyte replication. We conclude that RV-mediated hepatic gene therapy is effective for treating hemophilia B in mice and dogs, although the immune system may complicate gene transfer in adults.


Gene Therapy ◽  
1999 ◽  
Vol 6 (2) ◽  
pp. 209-218 ◽  
Author(s):  
A-M Darquet ◽  
R Rangara ◽  
P Kreiss ◽  
B Schwartz ◽  
S Naimi ◽  
...  

1997 ◽  
Vol 71 (11) ◽  
pp. 8221-8229 ◽  
Author(s):  
T J Wickham ◽  
E Tzeng ◽  
L L Shears ◽  
P W Roelvink ◽  
Y Li ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 452-452
Author(s):  
Ou Cao ◽  
Lixin Wang ◽  
Sushrusha Nayak ◽  
Roland W. Herzog

Abstract Gene therapy for the X-linked bleeding disorder hemophilia B may be limited by immune responses to the factor IX (F.IX) gene product. Hepatic adeno-associated virus (AAV) gene transfer can induce immune tolerance to F.IX (JCI111:1347, PNAS103:4592). Tolerance is associated with activation of regulatory cells that suppress antibody formation to F.IX. In order to identify these regulatory cells, splenocytes of C57BL/6 mice tolerized to human F.IX (hF.IX) by heptic gene transfer (portal vein injection of 1x1011 AAV vector genomes) were adoptively transferred to naive mice of the same strain. Recipient mice were immunized with hF.IX in adjuvant on the next day. Compared to cells transferred from control animals (no gene transfer), total splenocytes, CD4+ cells, or CD4+CD25+ cells were equally efficient in suppression of anti-hF.IX formation (n=7–8 per experimental group, P&lt;0.02 for comparison to total splenocytes, CD4+ cells, or CD4+CD25- cells of controls), while CD4- cells failed to suppress, and CD4+CD25- cells were inefficient. CD4+CD25+ from naive control mice, which contain regulatory T cells but lack specificity for hF.IX, gave highly variable results and on average failed to suppress. When tolerized C57BL/6 mice were challenged with hF.IX/adjuvant, the animals lacked antibody formation to hF.IX and in vitro cytokine release and showed an ~2-fold increase in FoxP3 message in splenic CD4+ cells in vivo. Taken together, these data indicate that induction of regulatory CD4+CD25+ T cells is part of the tolerance mechanism. However, the significance of this finding was unclear. In the next experiment, C57BL/6 mice received hepatic AAV-hF.IX gene transfer and were additionally injected with rat anti-mouse CD25 or with isotype control rat IgG (ip injections at days 0, 14, 28, and 42, n=5 per group). Analysis of peripheral blood cells by flow cytometry showed presence of CD4+CD25+ cells at a frequency of 8–10% in controls and undetectable levels in anti-CD25 treated mice. By day 49, 4/5 anti-CD25 treated mice had a low-titer, but detectable antibody (IgG1) to hF.IX. Subsequent challenge with hF.IX/cF.IX caused a rise in anti-hF.IX to 0.5–2 μg/ml in 3/5 anti-CD25 treated mice within 3 weeks. None of the mice treated with control IgG (0/5) developed a detectable antibody to hF.IX even after challenge. These data demonstrate that CD4+CD25+ regulatory T cells are required for tolerance induction to F.IX. Thus far, we failed to break tolerance by depletion of CD25+ cells at later time points (i.e. during the maintenance phase of tolerance, when other mechanisms such as T cell anergy and deletion may become more prevalent). To obtain definitive evidence for induction of CD4+CD25+ Treg, hepatic AAV-ova gene transfer was performed in DO11.10-tg Rag-2 −/− BALB/c mice, which are deficient in Treg. The DO11.10 T cell receptor is specific for ova peptide 323–339/MHC class II I-Ad complex. Within 2 weeks after gene transfer, CD4+CD25+GITR+ cells emerged in the thymus and in secondary lymphoid organs. Frequency of these cells increased to 2–4% by 2 months and subsequently remained at that level. These cells also expressed CTLA-4 and FoxP3 (&gt;100-fold increase in FoxP3 message compared to CD4+ cells from naive mice or compared to CD4+CD25- cells of AAV-ova transduced mice), and efficiently suppressed CD4+CD25- cells in vitro. In summary, hepatic AAV gene transfer induces transgene product-specific CD4+CD25+ Treg, which suppress antibody formation to the transgene product and are required for tolerance induction. These results should have broad implications for in vivo gene transfer.


Sign in / Sign up

Export Citation Format

Share Document