TY - JOUR AB - Glutaric aciduria type I (GA1) is an inherited disorder caused by the enzymatic defect of glutaryl-CoA dehydrogenase in the lysine degradation pathway, characterized by the accumulation of toxic metabolites in the central nervous system. We reasoned that substrate reduction therapy targeting the alpha-Aminoadipic Semialdehyde Synthase (AASS), the first enzyme in the catabolism of lysine, could provide an attractive therapeutic alternative. We explored to reduce the expression of AASS by an artificial microRNA with AASS target sequences embedded in a miR-16 backbone (miR_AASS). We analyzed several delivery routes and AAV serotypes and evaluated the therapeutic efficacy of a systemic neonatal delivery of AAV9_miR_AASS in the Gcdh-/- mouse model of GA1. We detected dose-dependent miR-AASS expression and AASS inhibition in liver and striatum, the main tissues affected in GA1. Treatment with AAV9_miR_AASS in lysine overload challenged mice reduced the accumulation of neurotoxic metabolites, up to six months post-treatment in the striatum, prevented the neuropathological alterations and improved mouse survival. Our results show that AAV9_miR_AASS supports AASS-lowering as a potential gene therapy strategy for GA1. AU - Segur-Bailach, E.* AU - Mateu-Bosch, A.* AU - Bofill-De Ros, X.* AU - Parés, M.* AU - da Silva Buttkus, P. AU - Rathkolb, B. AU - Gailus-Durner, V. AU - Hrabě de Angelis, M. AU - Moeini, P.* AU - Gonzalez-Aseguinolaza, G.* AU - Tort, F.* AU - Ribes, A.* AU - van Karnebeek, C.D.M.* AU - García-Villoria, J.* AU - Fillat, C.* C1 - 75181 C2 - 57822 CY - 50 Hampshire St, Floor 5, Cambridge, Ma 02139 Usa SP - 4820-4833 TI - Therapeutic AASS inhibition by AAV-miRNA rescues glutaric aciduria type I severe phenotype in mice. JO - Mol. Ther. VL - 33 IS - 10 PB - Cell Press PY - 2025 SN - 1525-0016 ER - TY - JOUR AB - Cancer cachexia is a severe systemic wasting disease that negatively affects quality of life and survival in patients with cancer. To date, treating cancer cachexia is still a major unmet clinical need. We recently discovered the destabilization of the AMP-activated protein kinase (AMPK) complex in adipose tissue as a key event in cachexia-related adipose tissue dysfunction and developed an adeno-associated virus (AAV)-based approach to prevent AMPK degradation and prolong cachexia-free survival. Here, we show the development and optimization of a prototypic peptide, Pen-X-ACIP, where the AMPK-stabilizing peptide ACIP is fused to the cell-penetrating peptide moiety penetratin via a propargylic glycine linker to enable late-stage functionalization using click chemistry. Pen-X-ACIP was efficiently taken up by adipocytes, inhibited lipolysis, and restored AMPK signaling. Tissue uptake assays showed a favorable uptake profile into adipose tissue upon intraperitoneal injection. Systemic delivery of Pen-X-ACIP into tumor-bearing animals prevented the progression of cancer cachexia without affecting tumor growth and preserved body weight and adipose tissue mass with no discernable side effects in other peripheral organs, thereby achieving proof of concept. As Pen-X-ACIP also exerted its anti-lipolytic activity in human adipocytes, it now provides a promising platform for further (pre)clinical development toward a novel, first-in-class approach against cancer cachexia. AU - Ji, H. AU - Englmaier, F. AU - Morigny, P. AU - Giroud, M. AU - Gräsle, P. AU - Brings, S.* AU - Szendrödi, J.* AU - Berriel Diaz, M. AU - Plettenburg, O. AU - Herzig, S. AU - Rohm, M. C1 - 68390 C2 - 54624 CY - 50 Hampshire St, Floor 5, Cambridge, Ma 02139 Usa SP - 2408-2421 TI - Development of a peptide drug restoring AMPK and adipose tissue functionality in cancer cachexia. JO - Mol. Ther. VL - 31 IS - 8 PB - Cell Press PY - 2023 SN - 1525-0016 ER - TY - JOUR AB - CD19-directed chimeric antigen receptor (CAR) T-cells have yielded impressive response rates in refractory/relapse B-cell acute lymphoblastic leukemia (B-ALL);however, most patients ultimately relapse due to poor CAR T-cell persistence or resistance of either CD19+ or CD19- B-ALL clones. CD22 is a pan-B marker whose expression is maintained in both CD19+ and CD19- relapses. Indeed, CD22-CAR T-cells have been clinically used in B-ALL patients, although relapse also occurs. Tcells engineered with a tandem CAR (Tan-CAR) containing in a single contruct both CD19 and CD22 scFvs, might be advantageus in achieving higher remission rates and/or preventing antigen loss. We have generated and functionally validated using cutting-edge assays a 4-1BB-based CD22/CD19 Tan-CAR using in-house-developed novel CD19 and CD22 scFvs. Tan-CAR-expressing T-cells showed similar in vitro expansion than CD19-CAR T-cells with no increased of tonic signaling. CRISPR/Cas9-edited B-ALL cells confirmed the bispecificity of the Tan-CAR. Tan-CAR was as efficient as CD19-CAR in vitro and in vivo using B-ALL cell lines, patient samples and patient-derived xenografts (PDXs). Strikingly, the robust anti-leukemic activity of the Tan-CAR was slightly more effective in controling the disease in long-term follow-up PDX models. This Tan-CAR construct warrants a clinical appraisal to test whether simultaneous targeting of CD19 and CD22 enhances leukemia eradication and reduces/delays relapse rates and antigen loss. AU - Zanetti, S.R.* AU - Velasco-Hernandez, T.* AU - Gutiérrez-Agüera, F.* AU - Díaz, V.M.* AU - Romecín, P.A.* AU - Roca-Ho, H.* AU - Sánchez-Martínez, D.* AU - Tirado, N.* AU - Baroni, M.L.* AU - Petazzi, P.* AU - Torres-Ruiz, R.A.* AU - Molina, O.* AU - Bataller, A.* AU - Fuster, J.L.* AU - Ballerini, P.* AU - Juan, M.* AU - Jeremias, I. AU - Bueno, C.* AU - Menéndez, P.* C1 - 62958 C2 - 51207 SP - 550-563 TI - A novel and efficient tandem CD19- and CD22-directed CAR for B-cell ALL. JO - Mol. Ther. VL - 30 IS - 2 PY - 2022 SN - 1525-0016 ER - TY - JOUR AU - Schreiber, S. AU - Honz, M. AU - Schiemann, M.* AU - Sette, A.* AU - Zielinski, C. AU - Protzer, U. AU - Wisskirchen, K. C1 - 59283 C2 - 48714 CY - 50 Hampshire St, Floor 5, Cambridge, Ma 02139 Usa SP - 520-520 TI - Characterization of MHC class II-restricted t-cell receptors for t-cell therapy of HBV infection. JO - Mol. Ther. VL - 28 IS - 4 PB - Cell Press PY - 2020 SN - 1525-0016 ER - TY - JOUR AB - Chimeric antigen receptor (CAR) T cell therapy is a promising novel therapeutic approach for cancer but also for chronic infection. We have developed a fully human, second-generation CAR directed against the envelope protein of hepatitis B virus on the surface of infected cells (S-CAR). The S-CAR contains a human B cell-derived single-chain antibody fragment and human immunoglobulin G (IgG) spacer, CD28- and CD3-signaling domains that may be immunogenic in mice. Because immunosuppression will worsen the clinical course of chronic hepatitis B, we aimed at developing a preclinical mouse model that is immunocompetent and mimics chronic hepatitis B but nevertheless allows evaluating efficacy and safety of a fully human CAR. The S-CAR grafted on T cells triggered antibody responses in immunocompetent animals, and a co-expressed human-derived safeguard, the truncated epidermal growth factor receptor (EGFRt), even induced B and T cell responses, both limiting the survival of S-CAR-grafted T cells. Total body irradiation and transfer of T cells expressing an analogous, signaling-deficient S-CAR decoy and the safeguard induced immune tolerance toward the human-derived structures. S-CAR T cells transferred after immune recovery persisted and showed long-lasting antiviral effector function. The approach we describe herein will enable preclinical studies of efficacy and safety of fully human CARs in the context of a functional immune system. AU - Festag, M. AU - Festag, J. AU - Fräßle, S.P.* AU - Asen, T. AU - Sacherl, J. AU - Schreiber, S. AU - Mück-Häusl, M. AU - Busch, D.H.* AU - Wisskirchen, K. AU - Protzer, U. C1 - 55644 C2 - 46494 CY - 50 Hampshire St, Floor 5, Cambridge, Ma 02139 Usa SP - 947-959 TI - Evaluation of a fully human, hepatitis B virus-specific chimeric antigen receptor in an immunocompetent mouse model. JO - Mol. Ther. VL - 27 IS - 5 PB - Cell Press PY - 2019 SN - 1525-0016 ER - TY - JOUR AU - Kalbarczyk, M.* AU - Slabik, C.* AU - Danisch, S.* AU - Cornelius, A.* AU - Zeidler, R. AU - Kroenke, N.* AU - Feuerhake, F.* AU - Stripecke, R.* C1 - 55967 C2 - 46676 CY - 50 Hampshire St, Floor 5, Cambridge, Ma 02139 Usa SP - 175-176 TI - Cord blood derived CAR-T cells targeting gp350 containing CD28/CD3 zeta or 4-1BB/CD3 zeta signaling domains directly react against cells infected with EBV bypassing the need of HLA-matched memory T cells. JO - Mol. Ther. VL - 27 IS - 4 PB - Cell Press PY - 2019 SN - 1525-0016 ER - TY - JOUR AB - Malaria, caused by protozoan Plasmodium parasites, remains a prevalent infectious human disease due to the lack of an efficient and safe vaccine. This is directly related to the persisting gaps in our understanding of the parasite's interactions with the infected host, especially during the clinically-silent yet essential liver stage of Plasmodium development. Previously, we and others showed that genetically-attenuated parasites (GAP) that arrest in the liver induce sterile immunity, but only upon multiple administrations. Here, we comprehensively studied hepatic gene and miRNA expression in GAP-injected mice, and found both a broad activation of IFNγ-associated pathways and a significant increase of murine microRNA-155 (miR-155), that was especially pronounced in non-parenchymal cells including liver-resident macrophages (Kupffer cells). Remarkably, ectopic upregulation of this miRNA in the liver of mice using robust hepatotropic Adeno-associated virus 8 (AAV8) vectors enhanced GAP's protective capacity substantially. In turn, this AAV8-mediated miR-155 expression permitted a reduction of GAP injections needed to achieve complete protection against infectious parasite challenge from previously three to only one. Our study highlights a crucial role of mammalian miRNAs in Plasmodium liver infection in vivo and concurrently implies their great potential as future immune-augmenting agents in improved vaccination regimes against malaria and other diseases. AU - Hentzschel, F.* AU - Hammerschmidt-Kamper, C.* AU - Börner, K.* AU - Heiss, K.* AU - Knapp, B. AU - Sattler, J.M.* AU - Kaderali, L.* AU - Castoldi, M.* AU - Bindman, J.G.* AU - Malato, Y.* AU - Willenbring, H.* AU - Mueller, A.K.* AU - Grimm, D.* C1 - 32103 C2 - 34967 SP - 2130-2141 TI - AAV8-mediated in vivo overexpression of miR-155 enhances the protective capacity of genetically-attenuated malarial parasites. JO - Mol. Ther. VL - 22 IS - 12 PY - 2014 SN - 1525-0016 ER - TY - JOUR AB - Recombinant vesicular stomatitis virus (VSV) shows promise for the treatment of hepatocellular carcinoma (HCC), but its safety and efficacy when administered in a setting of hepatic fibrosis, which occurs in the majority of clinical cases, is unknown. We hypothesized that VSV could provide a novel benefit to the underlying fibrosis, due to its ability to replicate and cause cell death specifically in activated hepatic stellate cells. In addition to the ability of VSV to produce a significant oncolytic response in HCC-bearing rats in the background of thioacetamide-induced hepatic fibrosis without signs of hepatotoxicity, we observed a significant downgrading of fibrosis stage, a decrease in collagen content in the liver, and modulation of gene expression in favor of fibrotic regression. Together, this work suggests that VSV is not only safe and effective for the treatment of HCC with underlying fibrosis, but it could potentially be developed for clinical application as a novel antifibrotic agent. AU - Altomonte, J.* AU - Marozin, S.* AU - de Toni, E.N.* AU - Rizzani, A.* AU - Esposito, I. AU - Steiger, K. AU - Feuchtinger, A. AU - Hellerbrand, C.* AU - Schmid, R.M.* AU - Ebert, O.* C1 - 27656 C2 - 32788 SP - 2032-2042 TI - Antifibrotic properties of transarterial oncolytic VSV therapy for hepatocellular carcinoma in rats with thioacetamide-induced liver fibrosis. JO - Mol. Ther. VL - 21 IS - 11 PB - Nature Publishing PY - 2013 SN - 1525-0016 ER - TY - JOUR AB - A number of recent reports have demonstrated that attenuated Salmonella typhimurium are capable of targeting both primary and metastatic tumors. The use of bacteria as a vehicle for the delivery of anticancer drugs requires a mechanism that precisely regulates and visualizes gene expression to ensure the appropriate timing and location of drug production. To integrate these functions into bacteria, we used a repressor-regulated tetracycline efflux system, in which the expression of a therapeutic gene and an imaging reporter gene were controlled by divergent promoters (tetAP and tetRP) in response to extracellular tetracycline. Attenuated S. typhimurium was transformed with the expression plasmids encoding cytolysin A, a therapeutic gene, and renilla luciferase variant 8, an imaging reporter gene, and administered intravenously to tumor-bearing mice. The engineered Salmonella successfully localized to tumor tissue and gene expression was dependent on the concentration of inducer, indicating the feasibility of peripheral control of bacterial gene expression. The bioluminescence signal permitted the localization of gene expression from the bacteria. The engineered bacteria significantly suppressed both primary and metastatic tumors and prolonged survival in mice. Therefore, engineered bacteria that carry a therapeutic and an imaging reporter gene for targeted anticancer therapy can be designed as a theranostic agent. AU - Jiang, S.N.* AU - Park, S.H.* AU - Lee, H.J.* AU - Zheng, J.H.* AU - Kim, H.S.* AU - Bom, H.S.* AU - Hong, Y.* AU - Szardenings, M.* AU - Shin, M.G.* AU - Kim, S.C.* AU - Ntziachristos, V. AU - Choy, H.E.* AU - Min, J.J.* C1 - 27499 C2 - 32700 SP - 1985-1995 TI - Engineering of bacteria for the visualization of targeted delivery of a cytolytic anticancer agent. JO - Mol. Ther. VL - 21 IS - 11 PB - Nature Publishing PY - 2013 SN - 1525-0016 ER - TY - JOUR AB - Tumor necrosisfactor alpha (TNF alpha) is a potent antitumoral cytokine, either killing tumor cells directly or affecting the tumor vasculature leading to enhanced accumulation of macromolecular drugs. Due to dose limiting side effects systemic administration of TNF alpha protein at therapeutically active doses is precluded. With gene vectors, tumor restricted TNF alpha expression can be achieved and in principle synergize with chemotherapy. Synthetic gene carriers based on polyamines were intravenously injected, which either passively accumulate within the tumor or specifically target the epidermal growth factor receptor. A single intravenous injection of TNF alpha gene vector promoted accumulation of liposomal doxorubicine (Doxil) in murine neuroblastoma and human hepatoma by enhancing tumor endothelium permeability. The expression of transgenic TNF alpha was restricted to tumor tissue. Three treatment cycles with TNF alpha gene vectors and Doxil significantly delayed tumor growth in subcutaneous murine Neuro2A neuroblastoma. Also tumors re-growing after initial treatment were successfully treated in a fourth cycle pointing at the absence of resistance mechanisms. Systemic Neuro2A metastases or human LS174T colon carcinoma metastases in liver were also successfully treated with this combined approach. In conclusion, this schedule opens the possibility for the efficient treatment of tumors metastases otherwise not accessible for macromolecular drug carriers. AU - Su, B.W.* AU - Cengizeroglu, A.* AU - Farkasova, K.* AU - Viola, J.R.* AU - Anton, M.* AU - Ellwart, J.W. AU - Haase, R.* AU - Wagner, E.* AU - Ogris, M.* C1 - 23389 C2 - 31027 SP - 300-308 TI - Systemic TNFα gene therapy synergizes with liposomal doxorubicine in the treatment of metastatic cancer. JO - Mol. Ther. VL - 21 IS - 2 PB - Nature Publishing Group PY - 2013 SN - 1525-0016 ER - TY - JOUR AB - Induced pluripotent stem cells (iPSCs) can be derived from somatic cells by gene transfer of reprogramming transcription factors. Expression levels of these factors strongly influence the overall efficacy to form iPSC colonies, but additional contribution of stochastic cell-intrinsic factors has been proposed. Here, we present engineered color-coded lentiviral vectors in which codon-optimized reprogramming factors are co-expressed by a strong retroviral promoter that is rapidly silenced in iPSC, and imaged the conversion of fibroblasts to iPSC. We combined fluorescence microscopy with long-term single cell tracking, and used live-cell imaging to analyze the emergence and composition of early iPSC clusters. Applying our engineered lentiviral vectors, we demonstrate that vector silencing typically occurs prior to or simultaneously with the induction of an Oct4-EGFP pluripotency marker. Around 7 days post-transduction (pt), a subfraction of cells in clonal colonies expressed Oct4-EGFP and rapidly expanded. Cell tracking of single cell-derived iPSC colonies supported the concept that stochastic epigenetic changes are necessary for reprogramming. We also found that iPSC colonies may emerge as a genetic mosaic originating from different clusters. Improved vector design with continuous cell tracking thus creates a powerful system to explore the subtle dynamics of biological processes such as early reprogramming events. AU - Warlich, E.* AU - Kuehle, J.* AU - Cantz, T.* AU - Brugman, M.H.* AU - Maetzig, T.* AU - Galla, M.* AU - Filipczyk, A.A. AU - Halle, S.* AU - Klump, H.* AU - Schöler, H.R.* AU - Baum, C.* AU - Schroeder, T. AU - Schambach, A.* C1 - 6648 C2 - 29038 CY - San Diego, CA, USA SP - 782-789 TI - Lentiviral vector design and imaging approaches to visualize the early stages of cellular reprogramming. JO - Mol. Ther. VL - 19 IS - 4 PB - Academic Press PY - 2011 SN - 1525-0016 ER - TY - JOUR AU - Javorovic, M. AU - Pohla, H. AU - Frankenberger, B. AU - Wölfel, T.* AU - Schendel, D.J. C1 - 3364 C2 - 23308 SP - 734-743 TI - RNA transfer by electroporation into mature dendritic cells leading to reactivation of effector-memory cytotoxic T lymphocytes: A quantitative analysis. JO - Mol. Ther. VL - 12 PY - 2005 SN - 1525-0016 ER - TY - JOUR AU - Perabo, L.* AU - Buning, H.* AU - Kofler, D.M. AU - Ried, M.U.* AU - Girod, A.* AU - Wendtner, C.-M. AU - Enssle, J.* AU - Hallek, M. C1 - 22339 C2 - 21196 SP - 151-157 TI - In Vitro Selection of Viral Vectors with Modified Tropism : the Adeno-associated Virus Display. JO - Mol. Ther. VL - 8 PY - 2003 SN - 1525-0016 ER -