TY - JOUR AB - Mutations in Dystrophin, one of the largest proteins in the mammalian body, are causative for a severe form of muscle disease, Duchenne Muscular Dystrophy (DMD), affecting not only skeletal muscle, but also the heart. In particular, exons 45-52 constitute a hotspot for DMD mutations. A variety of molecular therapies have been developed, comprising vectors encoding micro- and minidystrophins as well as utrophin, a protein with partially overlapping functions. With the advent of the CRISPR-Cas9-nuclease, genome editing offers a novel option of correction of the disease-cuasing mutations. Full restoration of the healthy gene by homology directed repair is a rare event. However, non-homologous end-joining (NHEJ) may restore the reading frame by causing exon excision. This approach has first been demonstrated in mice and then translated to large animals (dogs, pigs). This review discusses the potential opportunities and limitations of genome editing in DMD, including the generation of appropriate animal models as well as new developments in genome editing tools. AU - Kupatt, C.* AU - Windisch, A.* AU - Moretti, A.* AU - Wolf, E.* AU - Wurst, W. AU - Walter, M.C.* C1 - 61184 C2 - 50074 CY - Campus, 4 Crinan St, London, N1 9xw, England SP - 542–548 TI - Genome editing for Duchenne muscular dystrophy: A glimpse of the future? JO - Gene Ther. VL - 28 PB - Springernature PY - 2021 SN - 0969-7128 ER - TY - JOUR AB - Immune responses against multiple epitopes are required for the prevention of hepatitis C virus (HCV) infection, and the progression to phase I trials of candidates may be guided by comparative immunogenicity studies in non-human primates. Four vectors, DNA, SFV, human serotype 5 adenovirus (HuAd5) and Modified Vaccinia Ankara (MVA) poxvirus, all expressing hepatitis C virus Core, E1, E2 and NS3, were combined in three prime-boost regimen, and their ability to elicit immune responses against HCV antigens in rhesus macaques was explored and compared. All combinations induced specific T-cell immune responses, including high IFN-gamma production. The group immunized with the SFV+MVA regimen elicited higher E2-specific responses as compared with the two other modalities, while animals receiving HuAd5 injections elicited lower IL-4 responses as compared with those receiving MVA. The IFN-gamma responses to NS3 were remarkably similar between groups. Only the adenovirus induced envelope-specific antibody responses, but these failed to show neutralizing activity. Therefore, the two novel regimens failed to induce superior responses as compared with already existing HCV vaccine candidates. Differences were found in response to envelope proteins, but the relevance of these remain uncertain given the surprisingly poor correlation with immunogenicity data in chimpanzees, underlining the difficulty to predict efficacy from immunology studies. AU - Rollier, C.S.* AU - Verschoor, E.J.* AU - Verstrepen, B.E.* AU - Drexhage, J.A.R.* AU - Paranhos-Baccala, G.* AU - Liljeström, P.* AU - Sutter, G. AU - Arribillaga, L.* AU - Lasarte, J.J.* AU - Bartosch, B.* AU - Cosset, F.L.* AU - Inchauspe, G.* AU - Heeney, J.L.* C1 - 49918 C2 - 41917 CY - London SP - 753-759 TI - T- and B-cell responses to multivalent prime-boost DNA and viral vectored vaccine combinations against hepatitis C virus in non-human primates. JO - Gene Ther. VL - 23 IS - 10 PB - Nature Publishing Group PY - 2016 SN - 0969-7128 ER - TY - JOUR AB - Gene therapy holds great potential for the treatment of various acquired and inherited pulmonary diseases. Among various viral vectors, adeno-associated viral (AAV) vectors have been most frequently used in different clinical trials of pulmonary gene therapy. In the present study, we examined the kinetics and duration of transgene expression, vector biodistribution and development of neutralizing antibodies (NAB) in mice after pulmonary application of AAV2/9 vector. The pulmonary route of application did not affect any of the measured parameters. Transgene expression and biodistribution analysis at day 450 post-application confirmed the systemic spread of the vector after pulmonary delivery. Using SPB(-/-) mice, the study shows that AAV2/9-mediated gene expression is influenced by animal gender but not mouse genotype and is insensitive to the presence of lung inflammation. Lower expression levels were observed in male compared with female mice, and transient immunosuppression with dexamethasone significantly reduced the development of NAB in both genders of mice. The study thus advances this serotype for further development and use as a therapeutic vector. AU - Pfeifer, C.* AU - Aneja, M.K. AU - Hasenpusch, G.* AU - Rudolph, C.* C1 - 47041 C2 - 40530 SP - 1034-1042 TI - Adeno-associated virus serotype 9-mediated pulmonary transgene expression: effect of mouse strain, animal gender and lung inflammation. JO - Gene Ther. VL - 18 IS - 11 PY - 2011 SN - 0969-7128 ER - TY - JOUR AB - Adeno-associated virus (AAV)-mediated gene replacement for lysosomal disorders have been spurred by the ability of some serotypes to efficiently transduce neurons in the brain and by the ability of lysosomal enzymes to cross-correct among cells. Here, we explored enzyme replacement therapy in a knock-out mouse model of congenital neuronal ceroid lipofuscinosis (NCL), the most severe of the NCLs in humans. The missing protease in this disorder, cathepsin D (CathD) has high levels in the central nervous system. This enzyme has the potential advantage for assessing experimental therapy in that it can be imaged using a near-infrared fluorescence (NIRF) probe activated by CathD. Injections of an AAV2/rh8 vector-encoding mouse CathD (mCathD) into both cerebral ventricles and peritoneum of newborn knock-out mice resulted in a significant increase in lifespan. Successful delivery of active CathD by the AAV2/rh8-mCathD vector was verified by NIRF imaging of mouse embryonic fibroblasts from knock-out mice in culture, as well as by ex vivo NIRF imaging of the brain and liver after gene transfer. These studies support the potential effectiveness and imaging evaluation of enzyme replacement therapy to the brain and other organs in CathD null mice via AAV-mediated gene delivery in neonatal animals. AU - Pike, L.S.* AU - Tannous, B.A.* AU - Deliolanis, N.C. AU - Hsich, G.* AU - Morse, D.* AU - Tung, C.H.* AU - Sena-Esteves, M.* AU - Breakefield, X.O.* C1 - 6526 C2 - 28909 SP - 1173-1178 TI - Imaging gene delivery in a mouse model of congenital neuronal ceroid lipofuscinosis. JO - Gene Ther. VL - 18 IS - 12 PB - Nature Publ. Group PY - 2011 SN - 0969-7128 ER - TY - JOUR AB - Despite novel targeted agents, prognosis of metastatic renal cell cancer (RCC) remains poor, and experimental therapeutic strategies are warranted. Transfection of tumor cells with co-stimulatory molecules and/or cytokines is able to increase immunogenicity. Therefore, in our clinical study, 10 human leukocyte antigen (HLA)-A(*)0201(+) patients with histologically-confirmed progressive metastatic clear cell RCC were immunized repetitively over 22 weeks with 2.5-40 × 10(6) interleukin (IL)-7/CD80 cotransfected allogeneic HLA-A(*)0201(+) tumor cells (RCC26/IL-7/CD80). Endpoints of the study were feasibility, safety, immunological and clinical responses. Vaccination was feasible and safe. In all, 50% of the patients showed stable disease throughout the study; the median time to progression was 18 weeks. However, vaccination with allogeneic RCC26/IL-7/CD80 tumor cells was not able to induce TH1-polarized immune responses. A TH2 cytokine profile with increasing amounts of antigen-specific IL-10 secretion was observed in most of the responding patients. Interferon-γ secretion by patient lymphocytes upon antigen-specific and non-specific stimulation was substantially impaired, both before and during vaccination, as compared with healthy controls. This is possibly due to profound tumor-induced immunosuppression, which may prevent induction of antitumor immune responses by the gene-modified vaccine. Vaccination in minimal residual disease with concurrent depletion of regulatory cells might be one strategy to overcome this limitation. AU - Westermann, J.* AU - Flörcken, A.* AU - Willimsky, G.* AU - van, Lessen, A.* AU - Kopp, J.* AU - Takvorian, A.* AU - Jöhrens, K.* AU - Lukowsky, A.* AU - Schönemann, C.* AU - Sawitzki, B.* AU - Pohla, H. AU - Frank, R. AU - Dörken, B.* AU - Schendel, D.J. AU - Blankenstein, T.* AU - Pezzutto, A.* C1 - 6040 C2 - 28612 SP - 354-363 TI - Allogeneic gene-modified tumor cells (RCC-26/IL-7/CD80) as a vaccine in patients with metastatic renal cell cancer: A clinical phase-I study. JO - Gene Ther. VL - 18 IS - 4 PB - Nature Publ. Group PY - 2011 SN - 0969-7128 ER - TY - JOUR AB - Several vaccination trials are evaluating the modified vaccinia virus Ankara (MVA) as a delivery vector in various clinical settings. In this paper, we present the reevaluation of a therapeutic vaccination trial in human immunodeficiency virus (HIV)-1-infected individuals treated with highly active antiretroviral therapy using MVA-expressing HIV-1 nef. Immunogenicity of MVA-nef was assessed using multicolor flow cytometry. Vaccine-induced polyfunctionality and proliferative capacity, which are associated with nonprogressive HIV-1 infection, were detectable by combining two immune assays. By means of short-term polychromatic intracellular cytokine staining, we observed a significant increase in polyfunctional Nef-specific CD4 T cells expressing interferon-γ, interleukin (IL)-2 and CD154 after vaccination, whereas changes in the quality of CD8 T-cell response could not be observed. Only the additional use of a long-term polychromatic Carboxyfluorescein succinimidyl ester (CFSE)-based proliferation assay revealed vaccine-induced Nef-specific CD8, as well as CD4 T cells with proliferative capacity. The correlation between vaccine-induced IL-2 production by CD4 T cells and the increase in proliferating Nef-specific CD8 T cells suggests a causal link between these two functions. These results highlight the importance of combining sophisticated immunomonitoring tools to unravel concealed effects of immunological interventions and support the use of the poxvirus-derived MVA vector to stimulate highly functional HIV-1-specific T-cell responses. However, the clinical benefit of these functional T cells remains to be determined. AU - Kutscher, S. AU - Allgayer, S. AU - Dembek, C.J. AU - Bogner, J.R.* AU - Protzer, U. AU - Goebel, F.D.* AU - Erfle, V.* AU - Cosma, A. C1 - 4483 C2 - 27776 SP - 1372-1383 TI - MVA-nef induces HIV-1-specific polyfunctional and proliferative T-cell responses revealed by the combination of short- and long-term immune assays. JO - Gene Ther. VL - 17 IS - 11 PB - Nature Publ. Group PY - 2010 SN - 0969-7128 ER - TY - JOUR AB - no Abstract AU - Riddell, S.R.* AU - Protzer, U. C1 - 4564 C2 - 28263 SP - 1191-1192 TI - Grafting chimeric antigen receptors to redirect T cells: Carving the CAR. JO - Gene Ther. VL - 17 IS - 10 PB - Nature Publ. Group PY - 2010 SN - 0969-7128 ER - TY - JOUR AB - RNA interference allows selective gene silencing, and is widely used for functional analysis of individual genes in vertebrate cells and represents an attractive therapeutic option for treating central nervous system diseases. However, growing evidence exists that the expression of short hairpin RNAs (shRNAs) can trigger cellular immune response resulting in unspecific cellular phenotypes and severe side effects. We found that lentiviral vector (LV)-mediated expression of shRNAs in primary cortical cultures resulted in strong expression of the interferon-stimulated gene oligoadenylate synthetase 1 (Oas1), which was accompanied by accelerated apoptosis and substantial net neuron loss. Modification of the shRNA construct by implementing features of the naturally occurring microRNA-30 (miR-30) precursor avoided Oas1 induction in transduced primary cultures, whereby modification of the passenger strand seems to be a crucial feature to circumvent interferon-stimulated gene expression. This work represents the first experimental study showing that an miR-30-based shRNA construct prevents Oas1 pathway associated off-target effects, which we consider as an essential prerequisite for shRNA use in future gene therapeutic approaches. AU - Bauer, M. AU - Kinkl, N. AU - Meixner, A. AU - Kremmer, E. AU - Riemenschneider, M.* AU - Förstl, H.* AU - Gasser, T.* AU - Ueffing, M. C1 - 4057 C2 - 25769 SP - 142-147 TI - Prevention of interferon-stimulated gene expression using microRNA-designed hairpins. JO - Gene Ther. VL - 16 IS - 1 PB - Nature Publ. Group PY - 2009 SN - 0969-7128 ER - TY - JOUR AB - Neural progenitor cells are potential vehicles for delivery of therapeutic agents into the brain. Differentiation-dependent promoters may be useful to target the therapeutic transgene expression to specific neural cell types. Here we explored the potential of vectors based on the foamy virus (FV) for genetic engineering of neural progenitor cells. We demonstrate that FV vectors can mediate stable long-term constitutive expression of the enhanced green fluorescent protein (EGFP) in neural progenitor cells. For differentiation-dependent gene expression, we constructed a FV vector with an internal expression cassette containing the human 2.2 kb promoter (Gfa2) of the astrocyte-specific glial fibrillary acidic protein (GFAP) and sequences encoding EGFP. We show FV-vector-mediated delivery of the Gfa2-egfp transgene into the human neural stem cell line HNSC.100 and differentiation-dependent expression in stably transduced cell populations. Differentiation of the FV-transduced HNSC.100 cells to astrocytes upregulated expression of both the Gfa2-egfp transgene and the native gfap gene, confirming differentiation-dependent activation of the transduced Gfa2 promoter. These results demonstrate that differentiation-dependent gene expression can be achieved by FV-vector-mediated gene transfer to neural progenitor cells. Our findings support the use of FV vectors for the genetic engineering of neural progenitor cells for therapeutic and research applications. AU - Rothenaigner, I. AU - Kramer, S. AU - Meggendorfer, M. AU - Rethwilm, A.* AU - Brack-Werner, R. C1 - 3496 C2 - 25796 SP - 349-358 TI - Transduction of human neural progenitor cells with foamy virus vectors for differentiation-dependent gene expression. JO - Gene Ther. VL - 16 IS - 3 PB - Nature Publ. Group PY - 2009 SN - 0969-7128 ER - TY - JOUR AU - Hellebrand, E. AU - Mautner, J. AU - Reisbach, G. AU - Nimmerjahn, F. AU - Hallek, M.* AU - Mocikat, R. AU - Hammerschmidt, W. C1 - 4750 C2 - 23207 SP - 150-162 TI - Epstein-Barr virus vector-mediated gene transfer into human B cells: Potential for antitumor vaccination. JO - Gene Ther. VL - 13 PY - 2006 SN - 0969-7128 ER - TY - JOUR AU - Hettich, E. AU - Janz, A. AU - Zeidler, R.* AU - Pich, D. AU - Hellebrand, E. AU - Weissflog, B. AU - Moosmann, A. AU - Hammerschmidt, W. C1 - 5330 C2 - 23942 SP - 844-856 TI - Genetic design of an optimized packaging cell line for gene vectors transducing human B cells. JO - Gene Ther. VL - 13 PY - 2006 SN - 0969-7128 ER - TY - JOUR AU - Kofler, D.M. AU - Buning, H.* AU - Mayr, C. AU - Bund, D. AU - Baumert, J.J. AU - Hallek, M. AU - Wendtner, C.-M. C1 - 1903 C2 - 22401 SP - 1416-1424 TI - Engagement of the B-cell antigen receptor (BCR) allows efficient transduction of ZAP-70-positive primary B-CLL cells by recombinant adeno-associated virus (rAAV) vectors. JO - Gene Ther. VL - 11 PY - 2004 SN - 0969-7128 ER - TY - JOUR AU - Buning, H.* AU - Ried, M.U.* AU - Perabo, L.* AU - Gerner, F.M* AU - Huttner, N.A.* AU - Enssle, J.* AU - Hallek, M. C1 - 22338 C2 - 21195 SP - 1142-1151 TI - Receptor targeting of adeno-associated virus vectors. JO - Gene Ther. VL - 10 PY - 2003 SN - 0969-7128 ER - TY - JOUR AB - ven though renal cell carcinomas (RCC) are thought to be immunogenic, many tumors express variations in surface molecules and intracellular proteins that hinder induction of optimal antitumor responses. Interferon gamma (IFN gamma) stimulation can correct some of these deficiencies. Therefore, we introduced the complementary DNA (cDNA) encoding human IFN gamma into a well-characterized RCC line that has been selected for development of an allogeneic tumor cell vaccine for treatment of patients with metastatic disease. Studies were performed to determine how endogenous IFN gamma expression influences tumor cell immunogenicity. IFN gamma transductants showed minimal increases in surface expression of MHC class I and adhesion molecules but expression of class Ii molecules was induced. Proteins of the transporter associated with antigen processing (TAP) and low molecular weight polypeptide (LMP) were constitutively expressed at high levels. The transductants stimulated allospecific cytotoxic T lymphocytes (CTL); however, they were not better than unmodified tumor cells in this capacity. Endogenous IFN gamma expression enhanced tumor cell recognition by MHC-restricted, tumor antigen-specific CTL but suppressed recognition by non-MHC-restricted cytotoxic cells. Thus, the functional consequences of IFN gamma expression varied with respect to the type of effector cell and were not always beneficial for tumor cell recognition. AU - Schendel, D.J. AU - Falk, C.S. AU - Nößner, E. AU - Maget, B. AU - Kressenstein, S. AU - Urlinger, S.* AU - Tampé, R.* AU - Gansbacher, B.* C1 - 10168 C2 - 19345 SP - 2-10 TI - Gene transfer of human interferon gamma complementary DNA into a renal cell carcinoma line enhances MHC-restricted cytotoxic T lymphocyte recognition but suppresses non-MHC-restricted effector cell activity. JO - Gene Ther. VL - 7 PB - ature Publ. Group PY - 2000 SN - 0969-7128 ER - TY - JOUR AU - Selmayr, M. AU - Strehl, J. AU - Kremer, J.-P. AU - Kremmer, E. AU - Doenecke, A.* AU - Hallek, M.* AU - Menzel, H.* AU - Thielemans, K.* AU - Thierfelder, S. AU - Mocikat, R. C1 - 21230 C2 - 19329 SP - 778-784 TI - Induction of tumor immunity by autologous B lymphoma cells expressing a genetically engineered idiotype. JO - Gene Ther. VL - 6 PY - 1999 SN - 0969-7128 ER -