Paul J Rice-Boucher, Elena A Kashentseva, Igor P Dmitriev, Hongjie Guo, Jacqueline M Tremblay, Charles B Shoemaker, David T Curiel, Zhi Hong Lu
{"title":"<i>In-vivo</i>-targeted gene delivery using adenovirus-antibody site-specific covalent conjugates.","authors":"Paul J Rice-Boucher, Elena A Kashentseva, Igor P Dmitriev, Hongjie Guo, Jacqueline M Tremblay, Charles B Shoemaker, David T Curiel, Zhi Hong Lu","doi":"10.1016/j.omtm.2025.101497","DOIUrl":"10.1016/j.omtm.2025.101497","url":null,"abstract":"<p><p>Safe and efficient nucleic acid delivery to targeted cell populations remains a challenge in the fields of cell and gene therapy. Toward this end, we attempted to utilize the \"DogTag-DogCatcher\" system to target adenoviral vectors. \"DogTag\" is a short peptide that forms a spontaneous isopeptide bond upon mixing with its partner protein, \"DogCatcher.\" We genetically incorporated the DogTag peptide into the protein responsible for initial binding of the virus to its target cell, the fiber. This allowed permanent linking of DogCatcher-fused single-domain or single-chain antibodies at the fiber. This modification allowed simple, effective, and exclusive targeting of the vector to cells bound by the linked antibody. This enhanced gene transfer into primary B and T cells by up to 60-fold <i>in vitro</i> and 2- to 3-fold <i>in vivo</i> in mice without other alterations to vector tropism. Although the system's <i>in vivo</i> performance is currently suboptimal and additional engineering is needed prior to further use, these studies form the basis of a novel method for targeting adenovirus that can be combined with additional well-characterized adenovirus modifications toward applications in cell engineering, gene therapy, vaccines, oncolytics, and others.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101497"},"PeriodicalIF":4.6,"publicationDate":"2025-05-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12169769/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144310820","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
J P McGinnis, Joshua Ortiz-Guzman, Maria Camila Guevara, Sai Mallannagari, Benjamin D W Belfort, Suyang Bao, Snigdha Srivastava, Maria Morkas, Emily Ji, Angela Addison, Evelyne K Tantry, Sarah Chen, Ying Wang, Zihong Chen, Kalman A Katlowitz, Jeffrey J Lange, Melissa M Blessing, Carrie A Mohila, M Cecilia Ljungberg, Guillermo Aldave, Ali Jalali, Akash Patel, Sameer A Sheth, Howard L Weiner, Shankar Gopinath, Ganesh Rao, Akdes Serin Harmanci, Daniel J Curry, Benjamin R Arenkiel
{"title":"Common AAV gene therapy vectors show nonselective transduction of <i>ex vivo</i> human brain tissue.","authors":"J P McGinnis, Joshua Ortiz-Guzman, Maria Camila Guevara, Sai Mallannagari, Benjamin D W Belfort, Suyang Bao, Snigdha Srivastava, Maria Morkas, Emily Ji, Angela Addison, Evelyne K Tantry, Sarah Chen, Ying Wang, Zihong Chen, Kalman A Katlowitz, Jeffrey J Lange, Melissa M Blessing, Carrie A Mohila, M Cecilia Ljungberg, Guillermo Aldave, Ali Jalali, Akash Patel, Sameer A Sheth, Howard L Weiner, Shankar Gopinath, Ganesh Rao, Akdes Serin Harmanci, Daniel J Curry, Benjamin R Arenkiel","doi":"10.1016/j.omtm.2025.101494","DOIUrl":"10.1016/j.omtm.2025.101494","url":null,"abstract":"<p><p>The ability to deliver a therapeutic sequence to a specific cell type in the human brain would make possible innumerable therapeutic options for some of our most challenging diseases; however, studies on adeno-associated virus (AAV) vector tropism have generally relied on animal models with limited translational utility. For this reason, establishing the tropism of common adeno-associated virus (AAV) vectors in living human brain tissue serves as an important baseline for further optimization, as well as a determination of human brain cell types transduced by clinically approved gene therapy vectors AAV2 and AAV9. We have adapted an <i>ex vivo</i> organotypic model to evaluate AAV transduction properties in living slices of human brain tissue. Using fluorescent reporter expression and single-nucleus RNA sequencing, we found that common AAV vectors show broad transduction of normal cell types, with protein expression most apparent in astrocytes; this work introduces a pipeline for identifying and optimizing AAV gene therapy vectors in human brain samples.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101494"},"PeriodicalIF":4.6,"publicationDate":"2025-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12169722/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144310821","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yulia Eygeris, Antony Jozic, Michael I Henderson, Dylan Nelson, Gaurav Sahay
{"title":"Exploring the potential of saponins as adjuvants in lipid-nanoparticle-based mRNA vaccines.","authors":"Yulia Eygeris, Antony Jozic, Michael I Henderson, Dylan Nelson, Gaurav Sahay","doi":"10.1016/j.omtm.2025.101495","DOIUrl":"10.1016/j.omtm.2025.101495","url":null,"abstract":"<p><p>Saponins are a class of phytocompounds known for their amphiphilic properties. Here, we have evaluated incorporation of 40 saponins into a model lipid nanoparticle (LNP) formulation and evaluated their performance <i>in vitro</i> and <i>in vivo</i>. We reasoned that the surfactant activity of saponins could be beneficial in the context of cell and gene therapy due to the disruption of the intracellular membranes. We established formulation methodology to incorporate saponins into LNPs and measured their endosomal disruption and transfection efficiency with DNA barcode and mRNA cargoes. We identified two saponins-quillaic acid and macranthoidin B-that increase the LNP transfection efficiency and endosomal disruption. Saponin formulations demonstrated cargo-dependent activation of the innate immune system, as measured by the cell-based assays of interferon regulatory factor (IRF) and NF-κB pathway activation. Quillaic acid LNPs resulted in higher titers of anti-OVA IgG2a in the vaccination studies compared to a \"naive\" LNP control, which suggests a more Th1-biased immunopathology of these vaccines. As Th2-biased vaccines can trigger an allergic response, an mRNA vaccine with a balanced Th1/Th2 response is more favorable for translation into the clinic. Overall, quillaic acid may serve as an adjuvant for mRNA vaccines and potentially decrease the risk of vaccine-associated adverse events.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101495"},"PeriodicalIF":4.6,"publicationDate":"2025-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12169741/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144310822","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Adrien Soula, Florian Leseigneur, Amna Anwar, Bilal Ozdoganoglu, Jagan Gurung, Hamza Bhatti, Juline Guenat, Quentin Bazot, Majahar Sayed, Carolina Pinto Ricardo, Lily Li, Katerina Farukshina, Tony Bou Kheir, Hadi Mirmalek-Sani, Gregory Berger, Julie Kerby, Jonathan Appleby, Michael Delahaye
{"title":"The manufacture of AAV for gene therapy applications using a closed, semi-automated hollow-fiber bioreactor.","authors":"Adrien Soula, Florian Leseigneur, Amna Anwar, Bilal Ozdoganoglu, Jagan Gurung, Hamza Bhatti, Juline Guenat, Quentin Bazot, Majahar Sayed, Carolina Pinto Ricardo, Lily Li, Katerina Farukshina, Tony Bou Kheir, Hadi Mirmalek-Sani, Gregory Berger, Julie Kerby, Jonathan Appleby, Michael Delahaye","doi":"10.1016/j.omtm.2025.101496","DOIUrl":"10.1016/j.omtm.2025.101496","url":null,"abstract":"<p><p>Adeno-associated viral (AAV) vectors have been established as a safe and effective delivery vehicle for gene therapy. However, current methods for AAV production using adherent approaches are suboptimal due to their reliance on a substantial number of plastic-based flasks, manual labor, and a significant manufacturing footprint. Consequently, a protocol for generating AAV2 was developed on the Quantum, a semi-automated closed hollow-fiber bioreactor platform. In this system, Human Embryonic Kidney 293T cells were successfully expanded and transfected to produce an average crude AAV2 titer of 4.92 × 10<sup>14</sup> viral particles and 6.81 × 10<sup>13</sup> viral genomes from 1.2 L of harvested cell lysate. The application of a standard AAV downstream process confirmed normal processability of the material. A cost of goods model comparing the Quantum bioreactor with the current standard HYPERStack36 and Corning CellSTACK 10-layer systems demonstrated that the Quantum bioreactor reduced the number of open steps by more than 40-fold, production time by up to 3.6-fold (HYPERStack36) and 7.5-fold (CellSTACK 10-layer), and costs by up to 2-fold (HYPERStack36) and 20.7-fold (CellSTACK 10-layer). Therefore, the Quantum bioreactor is an effective alternative to plastic flasks for the manufacturing of AAVs at both R&D and early translational scale, as it reduces production time, operating costs, and process risk.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101496"},"PeriodicalIF":4.6,"publicationDate":"2025-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12167053/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144303586","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Christodoulos Messios, Andrie Koutsoulidou, Leonidas A Phylactou
{"title":"Widespread tissue delivery of antagomiRs via intramuscular administration.","authors":"Christodoulos Messios, Andrie Koutsoulidou, Leonidas A Phylactou","doi":"10.1016/j.omtm.2025.101488","DOIUrl":"10.1016/j.omtm.2025.101488","url":null,"abstract":"<p><p>Muscles, traditionally recognized for their role in locomotion and breathing, also participate in tissue communication. Extracellular microRNAs (miRNA) have been identified as key players in intercellular and inter-organ communication in muscle and other tissues. We have previously shown that intramuscular administration of an antagomiR led to the repression of target miRNA in neighboring skeletal muscles. This study investigated whether antagomiRs could be delivered to distant muscle and other tissues following intramuscular administration. We designed antagomiRs targeting a muscle-specific miRNA, miR-133b; a ubiquitously expressed miRNA, miR-16; and a scrambled oligonucleotide. Although all sequences were detected in neighboring skeletal muscles and distant tissues following intramuscular administration, antagomiR-133b showed the highest accumulation and efficacy in various tissues. This is the first study to provide evidence that intramuscular administration of antagomiRs could be utilized to achieve efficient and widespread distribution in tissues. This in turn could form the basis for alternative future therapeutic approaches.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101488"},"PeriodicalIF":4.6,"publicationDate":"2025-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12166816/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144303587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sofie Meulewaeter, Margo De Velder, Diethard Reckelbus, Kevin Mwangi, Thomas Ehouarne, Ilke Aernout, Yanou Engelen, Fellanza Halimi, Isis Van Herteryck, Lobke De Bels, Valerie Redant, Louise De la Mane, Joline Ingels, Bo Coppens, Serge Van Calenbergh, Pieter Cornillie, Gabriële Holtappels, Benedicte Descamps, Daisy Vanrompay, Stefaan C De Smedt, Wim Van den Broeck, Bart Vandekerckhove, Mathias Devreese, Rein Verbeke, Ine Lentacker
{"title":"Preclinical toxicological assessment of an α-galactosylceramide-adjuvanted mRNA cancer vaccine in Wistar Han rats and domestic pigs.","authors":"Sofie Meulewaeter, Margo De Velder, Diethard Reckelbus, Kevin Mwangi, Thomas Ehouarne, Ilke Aernout, Yanou Engelen, Fellanza Halimi, Isis Van Herteryck, Lobke De Bels, Valerie Redant, Louise De la Mane, Joline Ingels, Bo Coppens, Serge Van Calenbergh, Pieter Cornillie, Gabriële Holtappels, Benedicte Descamps, Daisy Vanrompay, Stefaan C De Smedt, Wim Van den Broeck, Bart Vandekerckhove, Mathias Devreese, Rein Verbeke, Ine Lentacker","doi":"10.1016/j.omtm.2025.101493","DOIUrl":"10.1016/j.omtm.2025.101493","url":null,"abstract":"<p><p>Galsome-NEO is a glycolipid-adjuvanted mRNA lipid nanoparticle (LNP) cancer vaccine encoding neo-epitopes for evaluation in a phase 1 study in patients with non-small cell lung cancer. To assess the safety of Galsome-NEO, a repeated-dose toxicity study was conducted in Wistar Han rats involving three intramuscular doses of 30 μg mRNA. A dose-escalation study in piglets tested three doses of 3, 15, and 100 μg mRNA. Rats showed a pronounced pro-inflammatory response, evidenced by cytokine secretion and an acute phase reaction. Clinical findings included temporary local reactions (maximum grade 3), elevated temperatures, and weight loss. In pigs, all doses were well tolerated. Blood analysis showed elevated alkaline phosphatase and decreased thrombocytes in rats, while pigs had reduced reticulocyte counts. Histology revealed hepatocyte vacuolation in rats and immune infiltration at injection sites in both species. In rats, blood and histology alterations resolved 3 weeks post dosing, except for immune infiltration in the connective tissue at injection sites in two females. Galsomes with mRNA encoding the <i>Chlamydia trachomatis</i> major outer membrane protein induced T cell responses in pigs. Natural killer T cell activation was observed in both species. These findings align with the safety data for the COVID-19 mRNA vaccine, Comirnaty, and demonstrate Galsomes' potential in large animals.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101493"},"PeriodicalIF":4.6,"publicationDate":"2025-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12167025/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144303584","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Qi Guo, Keval Parikh, Jian Zhang, Alexander Brinkley, Grace Chen, Natnicha Jakramonpreeya, Anjie Zhen, Dong Sung An
{"title":"Anti-HIV-1 HSPC-based gene therapy with safety kill switch to defend against and attack HIV-1 infection.","authors":"Qi Guo, Keval Parikh, Jian Zhang, Alexander Brinkley, Grace Chen, Natnicha Jakramonpreeya, Anjie Zhen, Dong Sung An","doi":"10.1016/j.omtm.2025.101486","DOIUrl":"10.1016/j.omtm.2025.101486","url":null,"abstract":"<p><p>Hematopoietic stem/progenitor cell (HSPC)-based anti-HIV-1 gene therapy holds promise to provide life-long remission following a single treatment. Here we report a multi-pronged anti-HIV-1 HSPC-based gene therapy designed to defend against and attack HIV-1 infection. We developed a lentiviral vector capable of co-expressing three anti-HIV-1 genes. Two are designed to prevent infection, including a short hairpin RNA (shRNA) (CCR5sh1005) to knock down HIV-1 co-receptor CCR5 and a membrane-anchored HIV-1 fusion inhibitor (C46). The third gene is a CD4-based chimeric antigen receptor (CAR) designed to attack HIV-1-infected cells. Our vector also includes a non-signaling truncated human epidermal growth factor receptor (huEGFRt) which acts as a negative selection-based safety kill switch against transduced cells. Anti-HIV-1 vector-transduced human CD34<sup>+</sup> HSPC efficiently reconstituted multi-lineage human hematopoietic cells in humanized bone marrow/liver/thymus (huBLT) mice. HIV-1 viral load was significantly reduced (1-log fold reduction, <i>p</i> < 0.001) in transplanted huBLT mice. Anti-huEGFR monoclonal antibody cetuximab (CTX) administration significantly reduced huEGFRt<sup>+</sup> vector-modified cells (>4-fold reduction, <i>p</i> < 0.01) in huBLT mice. These results demonstrate that our strategy is highly effective for HIV-1 inhibition, and that CTX-mediated negative selection can deplete anti-HIV-1 vector-modified cells in the event of unwanted adverse effects in huBLT mice.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101486"},"PeriodicalIF":4.6,"publicationDate":"2025-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12152878/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144276651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Junsik J Sung, Jacob R Shaw, Josie D Rezende, Shruti Dharmaraj, Andrea L Cottingham, Mehari M Weldemariam, Jace W Jones, Maureen A Kane, Ryan M Pearson
{"title":"Lipid nanoparticles from <i>L. meyenii</i> Walp mitigate sepsis through multimodal protein corona formation.","authors":"Junsik J Sung, Jacob R Shaw, Josie D Rezende, Shruti Dharmaraj, Andrea L Cottingham, Mehari M Weldemariam, Jace W Jones, Maureen A Kane, Ryan M Pearson","doi":"10.1016/j.omtm.2025.101491","DOIUrl":"10.1016/j.omtm.2025.101491","url":null,"abstract":"<p><p>Plant-derived lipid nanoparticles (PDNPs) are nano-sized particles isolated from various edible plants that contain bioactive components involved in regulating biological responses. Here, we isolated maca-derived lipid nanoparticles (MDNPs) from <i>Lepidium meyenii</i> Walp (maca), evaluated their therapeutic effects using two representative lethal models of sepsis, and determined their multimodal anti-inflammatory mechanism that relied on broad sequestration and neutralization of multiple pro-inflammatory cytokines and acute phase proteins (APPs) through formation of a protein corona. Lipidomics of MDNPs revealed triacylglycerols and phytoceramides as major constituents. <i>In vitro</i> studies showed that MDNPs were non-toxic, reduced macrophage activation, and sequestered lipopolysaccharide (LPS)-induced pro-inflammatory cytokines, while mitigating nuclear factor kappa B (NF-κB) activity. In a pre-established LPS-induced endotoxemia model, MDNP treatment significantly reduced systemic pro-inflammatory cytokines, reduced organ damage, and increased survival. Untargeted proteomics and bioinformatics analysis identified an enrichment in APPs present in MDNP protein coronas and corresponding inflammatory pathways modulated. The efficacy of MDNPs were further tested using a lethal polymicrobial sepsis model, where treatment significantly improved survival even in the absence of antibiotics. This study identifies MDNPs as an effective strategy capable of inducing potent anti-inflammatory responses, offering significant therapeutic potential for diseases such as sepsis, while informing the future design of synthetic lipid nanoparticles.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101491"},"PeriodicalIF":4.6,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12151679/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144267846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chloé Fustinoni, Xiang Liu, Zhi Chen, Ryan Cawood, Weimin Valenti, Maria I Patrício, Weiheng Su
{"title":"Self-silencing adenovirus enables precise infectious titration of recombinant adeno-associated viral vectors.","authors":"Chloé Fustinoni, Xiang Liu, Zhi Chen, Ryan Cawood, Weimin Valenti, Maria I Patrício, Weiheng Su","doi":"10.1016/j.omtm.2025.101492","DOIUrl":"10.1016/j.omtm.2025.101492","url":null,"abstract":"<p><p>Robust and accurate quantification of recombinant adeno-associated virus (rAAV) vectors' infectivity is essential for pre-clinical and clinical development of AAV gene therapy programs. The industry standard method for rAAV titration is the 50% tissue culture infectious dose (TCID50) assay using HeLa-based cell lines that stably encode the <i>rep</i> and <i>cap</i> genes from AAV serotype 2. Co-infection with wild-type (WT) adenoviruses provides the helper functions for expression of these genes, and the use of quantitative PCR (qPCR)/droplet digital PCR (ddPCR) serves as the endpoint method for the detection of infectious events. However, TCID50 assays using these HeLa-based <i>rep cap trans</i>-complementing cell lines have traditionally been regarded as challenging due to high variability, stability of the integrated genes, and safety concerns associated with the use of WT helper viruses. Here we developed a novel method for infectious titration of rAAV using our vector \"tetracycline-enabled self-silencing adenovirus\" (TESSA); we engineered it to deliver and express the AAV2 <i>rep</i> genes and adenoviral helper functions for rAAV genome replication, independent of the cell type. This approach allows the infectious titration of rAAV serotypes in cell lines permissive to adenovirus but without the production of adenoviral particles for improved safety, therefore benefiting GMP analytical requirements for rAAV gene therapies.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101492"},"PeriodicalIF":4.6,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12166451/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144303585","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anna Chen, Viktoria Knöbl, Oliver Walzer, Jana Hauser, Ines Neuwirth, Magdalena Frank, Nina Braun, Semina Duvnjak, Johannes Reisecker, Carmen Stecher, Alex Farr, Christine Brostjan, Dietmar Herndler-Brandstetter
{"title":"Comparison of NSG-Quad and MISTRG-6 humanized mice for modeling circulating and tumor-infiltrating human myeloid cells.","authors":"Anna Chen, Viktoria Knöbl, Oliver Walzer, Jana Hauser, Ines Neuwirth, Magdalena Frank, Nina Braun, Semina Duvnjak, Johannes Reisecker, Carmen Stecher, Alex Farr, Christine Brostjan, Dietmar Herndler-Brandstetter","doi":"10.1016/j.omtm.2025.101487","DOIUrl":"10.1016/j.omtm.2025.101487","url":null,"abstract":"<p><p>Humanized mice are valuable preclinical models for immuno-oncology research because they allow modeling of human immune cells and human tumors <i>in vivo</i>. Myeloid cells are highly abundant in many tumors and have been associated with tumor progression, metastasis, and therapy resistance. Next-generation humanized mice have been generated to improve the development, diversity, and function of human myeloid cells. In this study, we analyzed human immune cell development and myeloid cell composition in NSG-Quad and MISTRG-6 mice. NSG-Quad mice supported the development of tissue-resident and tumor-infiltrating human macrophages at levels almost comparable to those of MISTRG-6 mice. However, the development of human CD4<sup>+</sup> and CD8<sup>+</sup> T cells was impaired in the blood and spleen but not in the tumor of NSG-Quad mice. In a subset of NSG-Quad mice, human monocytes exhibited increased cellular granularity and elevated expression of activation and checkpoint molecules, consistent with a monocyte hyperactivation syndrome. Our study provides a comprehensive comparative analysis of the frequency and characteristics of circulating, tissue-resident, and tumor-infiltrating myeloid cell populations in NSG-Quad and MISTRG-6 mice, which is key to accurately design and interpret human tumor xenograft studies, particularly with regard to faithful reconstruction of the human tumor-immune microenvironment and preclinical testing.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101487"},"PeriodicalIF":4.6,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12152875/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144276652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}