Pub Date : 2025-05-19eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101493
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
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 Chlamydia trachomatis 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.
{"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}
Pub Date : 2025-05-15eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101486
Qi Guo, Keval Parikh, Jian Zhang, Alexander Brinkley, Grace Chen, Natnicha Jakramonpreeya, Anjie Zhen, Dong Sung An
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+ 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, p < 0.001) in transplanted huBLT mice. Anti-huEGFR monoclonal antibody cetuximab (CTX) administration significantly reduced huEGFRt+ vector-modified cells (>4-fold reduction, p < 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.
{"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}
Pub Date : 2025-05-14eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101491
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
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 Lepidium meyenii 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. In vitro 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.
{"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}
Pub Date : 2025-05-14eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101492
Chloé Fustinoni, Xiang Liu, Zhi Chen, Ryan Cawood, Weimin Valenti, Maria I Patrício, Weiheng Su
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 rep and cap 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 rep cap trans-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 rep 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.
{"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}
Pub Date : 2025-05-14eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101487
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
Humanized mice are valuable preclinical models for immuno-oncology research because they allow modeling of human immune cells and human tumors in vivo. 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+ and CD8+ 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.
{"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}
Pub Date : 2025-05-14eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101489
Elizabeth J Bealer, Namit Padgaonkar, Kelly Crumley, Eiji Saito, Zoe Beekman, Alexa DeKorte, Thazha P Prakash, Alexey Revenko, Lonnie D Shea
One of the many challenges for islet transplantation as a treatment for type 1 diabetes is inflammation that contributes to islet de-differentiation and death. Innate immune cells such as monocytes and macrophages secrete tumor necrosis factor alpha (TNF-α), interleukin 1β (IL-1β), inducible nitric oxide synthase (iNOS), and IL-6, which directly contribute to islet dysfunction. Attenuation of the early inflammatory response post-transplantation may protect cell survival and subsequent function. Herein, we investigate the development of anti-TNF-α antisense-oligonucleotide-conjugated polylactide-co-glycolide nanoparticles (PLG-aTNF-α NPs) as an anti-inflammatory therapy after stem-cell-derived islet transplantation. PLG-aTNF-α NPs are shelf stable and successfully reduce TNF-α secretion and expression in inflammatory macrophages. Synergy between the aTNF-α antisense oligonucleotide and the polylactide-co-glycolide NPs results in further knockdown of IL-1β, IL-6, iNOS, and IL-12 in vitro indicating PLG-aTNF-α NPs may protect against the inflammatory cascade in vivo. In a diabetic mouse model, stem-cell-derived islets transplanted to the peritoneal fat were protected after treatment with PLG-aTNF-α NPs compared with PLG NPs alone. Tnfα and Il1β expression was reduced in mice treated with PLG-aTNF-α NPs, indicating inflammation was reduced after transplant. PLG-aTNF-α NPs reduce TNF-α and protect islets, supporting their potential use as a therapeutic in islet transplantation.
{"title":"Anti-TNF-α antisense-oligonucleotide-conjugated PLG nanoparticles protect transplanted islets.","authors":"Elizabeth J Bealer, Namit Padgaonkar, Kelly Crumley, Eiji Saito, Zoe Beekman, Alexa DeKorte, Thazha P Prakash, Alexey Revenko, Lonnie D Shea","doi":"10.1016/j.omtm.2025.101489","DOIUrl":"10.1016/j.omtm.2025.101489","url":null,"abstract":"<p><p>One of the many challenges for islet transplantation as a treatment for type 1 diabetes is inflammation that contributes to islet de-differentiation and death. Innate immune cells such as monocytes and macrophages secrete tumor necrosis factor alpha (TNF-α), interleukin 1β (IL-1β), inducible nitric oxide synthase (iNOS), and IL-6, which directly contribute to islet dysfunction. Attenuation of the early inflammatory response post-transplantation may protect cell survival and subsequent function. Herein, we investigate the development of anti-TNF-α antisense-oligonucleotide-conjugated polylactide-co-glycolide nanoparticles (PLG-aTNF-α NPs) as an anti-inflammatory therapy after stem-cell-derived islet transplantation. PLG-aTNF-α NPs are shelf stable and successfully reduce TNF-α secretion and expression in inflammatory macrophages. Synergy between the aTNF-α antisense oligonucleotide and the polylactide-co-glycolide NPs results in further knockdown of IL-1β, IL-6, iNOS, and IL-12 <i>in vitro</i> indicating PLG-aTNF-α NPs may protect against the inflammatory cascade <i>in vivo</i>. In a diabetic mouse model, stem-cell-derived islets transplanted to the peritoneal fat were protected after treatment with PLG-aTNF-α NPs compared with PLG NPs alone. <i>Tnfα</i> and <i>I</i> <i>l</i> <i>1β</i> expression was reduced in mice treated with PLG-aTNF-α NPs, indicating inflammation was reduced after transplant. PLG-aTNF-α NPs reduce TNF-α and protect islets, supporting their potential use as a therapeutic in islet transplantation.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101489"},"PeriodicalIF":4.7,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12151673/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144267845","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}
Pub Date : 2025-05-14eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101490
Brett J G Estes, Nisha Gandhi, Jessica R Von Stetina, Dev Paudel, Angela X Nan, Parth Amin, Joshua Rose, Shuai Wu, Kangni Zheng, Yijun Zhang, Jesse C Cochrane, Jonathan D Finn, Jenny Xie
Recent advancements in gene insertion have shifted from DNA-repair-dependent mechanisms to more precise approaches, enhancing safety and predictability for editing outcomes. Integrase-mediated programmable genomic integration (I-PGI) utilizes a DNA cargo to insert transgenes in a targeted, unidirectional manner. In vivo, where nuclear delivery of DNA is challenging, adeno-associated virus (AAV) can act as the cargo vector. Although I-PGI does not require DNA double-strand breaks (DSBs) for activity, linear cargo, like AAV, stimulates DNA end-joining activity after integration. To mitigate potential risks from DSBs, we developed two circular AAV cargos capable of seamless gene insertion in non-dividing cells. We first harnessed the orthogonal property of large serine integrases to produce circle-AAV (cAAV) from linear viral genomes in cells. cAAV demonstrated seamless cargo integration in primary human hepatocytes (PHHs) and robust DSB-free insertion structures in vivo. We then investigated the delivery of a packaged circular AAV cargo (AAV.AD), which eliminates the need for enzymatic manipulation in the cell. AAV.AD exhibited functional seamless gene insertion in PHHs and showed cargo efficacy in vivo. Together, these findings provide evidence of DSB-free programmable genomic integration using integrase and AAV cargo, addressing a previously unrecognized challenge in the field.
{"title":"Development of circular AAV cargos for targeted seamless insertion with large serine integrases.","authors":"Brett J G Estes, Nisha Gandhi, Jessica R Von Stetina, Dev Paudel, Angela X Nan, Parth Amin, Joshua Rose, Shuai Wu, Kangni Zheng, Yijun Zhang, Jesse C Cochrane, Jonathan D Finn, Jenny Xie","doi":"10.1016/j.omtm.2025.101490","DOIUrl":"10.1016/j.omtm.2025.101490","url":null,"abstract":"<p><p>Recent advancements in gene insertion have shifted from DNA-repair-dependent mechanisms to more precise approaches, enhancing safety and predictability for editing outcomes. Integrase-mediated programmable genomic integration (I-PGI) utilizes a DNA cargo to insert transgenes in a targeted, unidirectional manner. <i>In vivo</i>, where nuclear delivery of DNA is challenging, adeno-associated virus (AAV) can act as the cargo vector. Although I-PGI does not require DNA double-strand breaks (DSBs) for activity, linear cargo, like AAV, stimulates DNA end-joining activity after integration. To mitigate potential risks from DSBs, we developed two circular AAV cargos capable of seamless gene insertion in non-dividing cells. We first harnessed the orthogonal property of large serine integrases to produce circle-AAV (cAAV) from linear viral genomes in cells. cAAV demonstrated seamless cargo integration in primary human hepatocytes (PHHs) and robust DSB-free insertion structures <i>in vivo</i>. We then investigated the delivery of a packaged circular AAV cargo (AAV.AD), which eliminates the need for enzymatic manipulation in the cell. AAV.AD exhibited functional seamless gene insertion in PHHs and showed cargo efficacy <i>in vivo</i>. Together, these findings provide evidence of DSB-free programmable genomic integration using integrase and AAV cargo, addressing a previously unrecognized challenge in the field.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101490"},"PeriodicalIF":4.6,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12166831/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144303583","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}
Pub Date : 2025-05-08eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101485
Junnan Shi, Jinagya Yang, Yu Zheng, Phyllis Hio Hong Wong, Hao Hu, Carolina Oi Lam Ung
Advanced therapy medicinal products (ATMPs) require rigorous quality management to mitigate risks associated with their development and use in hospitals. This study aimed to identify guidelines, standards of practice, and practical experiences in ATMPs quality management in hospital settings. An integrative scoping review under PRISMA guidelines retrieved 14 studies from four databases, 144 quality management guidelines and standards, and risk management reports for approved ATMPs from three government agencies and six organizations. Thirteen models or programs of quality management practices for ATMPs were identified across 25 hospital-based settings in six countries. Major aspects of ATMPs included clinical quality and translation, logistics management, hospital preparation, and patient care. Primary goals of ATMPs management within hospitals involved regulatory compliance and accreditation with national and regional requirements and implementing and maintaining the standardized operational practices. Four priority actions to enhance the quality of ATMPs management were as follows: (1) risk-based procedures and strategies; (2) strengthening of the skills and knowledge of healthcare professionals and technical staff; (3) validation and maintenance of qualified storage, manufacturing, and delivery facilities; and (4) support for a documentation system. In summary, understanding key components of ATMPs management offers valuable insights for developing an adaptive quality management ecosystem supporting clinical translation.
{"title":"The clinical quality management system of advanced therapy medicinal products in the hospital setting: A scoping review.","authors":"Junnan Shi, Jinagya Yang, Yu Zheng, Phyllis Hio Hong Wong, Hao Hu, Carolina Oi Lam Ung","doi":"10.1016/j.omtm.2025.101485","DOIUrl":"10.1016/j.omtm.2025.101485","url":null,"abstract":"<p><p>Advanced therapy medicinal products (ATMPs) require rigorous quality management to mitigate risks associated with their development and use in hospitals. This study aimed to identify guidelines, standards of practice, and practical experiences in ATMPs quality management in hospital settings. An integrative scoping review under PRISMA guidelines retrieved 14 studies from four databases, 144 quality management guidelines and standards, and risk management reports for approved ATMPs from three government agencies and six organizations. Thirteen models or programs of quality management practices for ATMPs were identified across 25 hospital-based settings in six countries. Major aspects of ATMPs included clinical quality and translation, logistics management, hospital preparation, and patient care. Primary goals of ATMPs management within hospitals involved regulatory compliance and accreditation with national and regional requirements and implementing and maintaining the standardized operational practices. Four priority actions to enhance the quality of ATMPs management were as follows: (1) risk-based procedures and strategies; (2) strengthening of the skills and knowledge of healthcare professionals and technical staff; (3) validation and maintenance of qualified storage, manufacturing, and delivery facilities; and (4) support for a documentation system. In summary, understanding key components of ATMPs management offers valuable insights for developing an adaptive quality management ecosystem supporting clinical translation.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101485"},"PeriodicalIF":4.6,"publicationDate":"2025-05-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12179598/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144477872","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}
Pub Date : 2025-05-07eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101484
Charles Moore-Kelly, Rajesh Reddem, Ben M Alberts, Jordan Wright, Thomas Evans, Anurag Kulkarni, Nicholas G Clarkson, Daniel C Farley, Kyriacos A Mitrophanous, Rui André Saraiva Raposo
Lentiviral vector (LV)-based therapies employ the molecular machinery of HIV-1 to stably integrate therapeutic genes into patient cells for long-term disease correction. However, suboptimal expression of LV components in HEK293T-based production systems can limit titers and hinder clinical product development. Here, we identify protein kinase C (PKC) agonists as robust enhancers of LV production. PKC activation resulted in rapid transcription of LV genomic RNA and accelerated vector particle release in a manner that complemented the use of the histone deacetylase (HDAC) inhibitor, sodium butyrate. Stimulation of HEK293T cells strongly upregulated AP-1 transcription factor subunits independently of nuclear factor κB (NF-κB) pathway activation. Application of PKC agonists in LV production resulted in a ∼3-fold improvement in the titer of a chimeric antigen receptor (CAR)-LV. Furthermore, a ∼9-fold increase in titer was achieved when this induction method was combined with co-expression of an LV RNA-targeted U1 snRNA enhancer. Importantly, LV produced using PKC agonists had comparable particle-to-infectivity ratios and preserved T cell transduction efficiency. These findings suggest that incorporating PKC agonists into commercial LV manufacturing could considerably reduce the cost per patient dose of new LV-based gene therapies.
{"title":"Enhancing titers of therapeutic lentiviral vectors using PKC agonists.","authors":"Charles Moore-Kelly, Rajesh Reddem, Ben M Alberts, Jordan Wright, Thomas Evans, Anurag Kulkarni, Nicholas G Clarkson, Daniel C Farley, Kyriacos A Mitrophanous, Rui André Saraiva Raposo","doi":"10.1016/j.omtm.2025.101484","DOIUrl":"10.1016/j.omtm.2025.101484","url":null,"abstract":"<p><p>Lentiviral vector (LV)-based therapies employ the molecular machinery of HIV-1 to stably integrate therapeutic genes into patient cells for long-term disease correction. However, suboptimal expression of LV components in HEK293T-based production systems can limit titers and hinder clinical product development. Here, we identify protein kinase C (PKC) agonists as robust enhancers of LV production. PKC activation resulted in rapid transcription of LV genomic RNA and accelerated vector particle release in a manner that complemented the use of the histone deacetylase (HDAC) inhibitor, sodium butyrate. Stimulation of HEK293T cells strongly upregulated AP-1 transcription factor subunits independently of nuclear factor κB (NF-κB) pathway activation. Application of PKC agonists in LV production resulted in a ∼3-fold improvement in the titer of a chimeric antigen receptor (CAR)-LV. Furthermore, a ∼9-fold increase in titer was achieved when this induction method was combined with co-expression of an LV RNA-targeted U1 snRNA enhancer. Importantly, LV produced using PKC agonists had comparable particle-to-infectivity ratios and preserved T cell transduction efficiency. These findings suggest that incorporating PKC agonists into commercial LV manufacturing could considerably reduce the cost per patient dose of new LV-based gene therapies.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101484"},"PeriodicalIF":4.6,"publicationDate":"2025-05-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12146000/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144259414","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}
Pub Date : 2025-04-30eCollection Date: 2025-06-12DOI: 10.1016/j.omtm.2025.101483
Raymond Wu, Frank Luh, Soo-Mi Kweon, Yun Yen
Viral copy-number (VCN) assay is a powerful, effective method to quantify toxicity, cellular kinetics, and durability of virus-modified cell therapy products. The qualification and validation of assay requires reference control. Traditionally, plasmids and cell lines are used as reference controls, but development and qualification of those controls require considerable time and resources. We propose a reference synthetic DNA fragment containing amplicons of woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) and ribonuclease P protein subunit p30 (RPP30), connected by HindIII restriction enzyme cutting site, as a useful tool to qualify and validate duplex droplet digital PCR (ddPCR) assays for VCN. Using this hybrid amplicon, we qualified the duplex WPRE/RPP30 ddPCR assay by determining range of quantification, precision, bias, and robustness of the assay. The varying amount of input DNA showed upper limit, lower limit, and linearity of the assay. Coefficient of variation (CV) and % recovery showed assay precision and accuracy, respectively. Furthermore, the hybrid amplicon was used to determine assay robustness with potential conditions of variability. The hybrid amplicon was a comparable alternative to cell reference standards for validating VCN assay. In conclusion, WPRE-RPP30 hybrid amplicon can be used as a routine quality control measure to validate digital PCR assays.
{"title":"A method to validate viral copy-number assay involving a hybrid amplicon and duplex droplet digital PCR.","authors":"Raymond Wu, Frank Luh, Soo-Mi Kweon, Yun Yen","doi":"10.1016/j.omtm.2025.101483","DOIUrl":"10.1016/j.omtm.2025.101483","url":null,"abstract":"<p><p>Viral copy-number (VCN) assay is a powerful, effective method to quantify toxicity, cellular kinetics, and durability of virus-modified cell therapy products. The qualification and validation of assay requires reference control. Traditionally, plasmids and cell lines are used as reference controls, but development and qualification of those controls require considerable time and resources. We propose a reference synthetic DNA fragment containing amplicons of woodchuck hepatitis virus posttranscriptional regulatory element (<i>WPRE</i>) and ribonuclease P protein subunit p30 (<i>RPP30</i>), connected by HindIII restriction enzyme cutting site, as a useful tool to qualify and validate duplex droplet digital PCR (ddPCR) assays for VCN. Using this hybrid amplicon, we qualified the duplex WPRE/RPP30 ddPCR assay by determining range of quantification, precision, bias, and robustness of the assay. The varying amount of input DNA showed upper limit, lower limit, and linearity of the assay. Coefficient of variation (CV) and % recovery showed assay precision and accuracy, respectively. Furthermore, the hybrid amplicon was used to determine assay robustness with potential conditions of variability. The hybrid amplicon was a comparable alternative to cell reference standards for validating VCN assay. In conclusion, <i>WPRE-RPP30</i> hybrid amplicon can be used as a routine quality control measure to validate digital PCR assays.</p>","PeriodicalId":54333,"journal":{"name":"Molecular Therapy-Methods & Clinical Development","volume":"33 2","pages":"101483"},"PeriodicalIF":4.6,"publicationDate":"2025-04-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12229726/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144576962","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}