Antibody gene transfer offers a promising solution to the high cost and frequent administration of monoclonal antibodies (mAbs), enabling the body to produce its own drugs economically and sustainably. This review addresses the challenges faced by antibody therapies, including economic and environmental impacts, as well as patient-related issues such as efficacy and tolerance. We propose that direct in vivo protein production, or autologous production, via plasmid DNA (pDNA) injection may address some of these challenges. This pDNA-based strategy provides a cost-effective alternative while maintaining flexibility and adaptability for various proteins, making it suitable for a wide range of pathological contexts. Additionally, gene therapy with plasmids could reduce the need for frequent injections, improving patient compliance. In this review, we provide an overview of the pioneering studies that introduced the use of pDNA for in vivo protein production. We focus on key factors for successful autologous production, such as plasmid design, vectorization, and methods of administration. Finally, we explore various applications where autologous production could serve as a promising alternative for therapeutic antibody treatments.
{"title":"Autologous Production: The Future of Sustainable Antibody Treatments.","authors":"Steevens Bouaziz, Florence Rouleux-Bonnin, Stéphanie David, Guillermo Carvajal Alegria, Florence Velge-Roussel","doi":"10.1089/hum.2025.052","DOIUrl":"10.1089/hum.2025.052","url":null,"abstract":"<p><p>Antibody gene transfer offers a promising solution to the high cost and frequent administration of monoclonal antibodies (mAbs), enabling the body to produce its own drugs economically and sustainably. This review addresses the challenges faced by antibody therapies, including economic and environmental impacts, as well as patient-related issues such as efficacy and tolerance. We propose that direct <i>in vivo</i> protein production, or autologous production, via plasmid DNA (pDNA) injection may address some of these challenges. This pDNA-based strategy provides a cost-effective alternative while maintaining flexibility and adaptability for various proteins, making it suitable for a wide range of pathological contexts. Additionally, gene therapy with plasmids could reduce the need for frequent injections, improving patient compliance. In this review, we provide an overview of the pioneering studies that introduced the use of pDNA for <i>in vivo</i> protein production. We focus on key factors for successful autologous production, such as plasmid design, vectorization, and methods of administration. Finally, we explore various applications where autologous production could serve as a promising alternative for therapeutic antibody treatments.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1222-1236"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144527693","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01DOI: 10.1177/10430342251372056
Jimena Pérez-Maroto, Laura Sepp-Lorenzino, Diego Castaño-Esteban, Daniela Palacios, Begoña Sot
Rare diseases are serious and often chronic conditions that affect a small number of individuals. However, with over 7,000 rare diseases identified, their cumulative global numbers and impact are substantial. A considerable proportion of these conditions is caused by genetic abnormalities. Among these, monogenic disorders are of particular relevance, as they are caused by mutations in specific genes. The development of gene therapy, and more specifically, gene editing, offers innovative approaches to treat these rare diseases. A significant challenge associated with the implementation of such strategies concerns the delivery of gene editing tools. Nonviral vectors based on nanomaterials have demonstrated considerable potential as promising alternatives to viral vectors, thereby overcoming their disadvantages. The biocompatibility and tunability of nanoparticles, along with their potential capacity to target diverse tissues, positions them as a promising therapeutic approach for the treatment of a wide range of organ-specific rare diseases. Here, we review current progress in the development and evaluation of novel nanomedicine strategies for gene editing in rare diseases, highlighting new gene editing approaches, delivery systems, and potential targets.
{"title":"Advancements in Nonviral Gene Editing Strategies for Rare Diseases.","authors":"Jimena Pérez-Maroto, Laura Sepp-Lorenzino, Diego Castaño-Esteban, Daniela Palacios, Begoña Sot","doi":"10.1177/10430342251372056","DOIUrl":"10.1177/10430342251372056","url":null,"abstract":"<p><p>Rare diseases are serious and often chronic conditions that affect a small number of individuals. However, with over 7,000 rare diseases identified, their cumulative global numbers and impact are substantial. A considerable proportion of these conditions is caused by genetic abnormalities. Among these, monogenic disorders are of particular relevance, as they are caused by mutations in specific genes. The development of gene therapy, and more specifically, gene editing, offers innovative approaches to treat these rare diseases. A significant challenge associated with the implementation of such strategies concerns the delivery of gene editing tools. Nonviral vectors based on nanomaterials have demonstrated considerable potential as promising alternatives to viral vectors, thereby overcoming their disadvantages. The biocompatibility and tunability of nanoparticles, along with their potential capacity to target diverse tissues, positions them as a promising therapeutic approach for the treatment of a wide range of organ-specific rare diseases. Here, we review current progress in the development and evaluation of novel nanomedicine strategies for gene editing in rare diseases, highlighting new gene editing approaches, delivery systems, and potential targets.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1118-1137"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144952013","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01Epub Date: 2025-08-22DOI: 10.1177/10430342251372047
Laura Rodríguez, Pablo de Felipe, Raquel Martín, Jorge Martínez, David Ordóñez, Susana Rojo, Juan Fernando Martínez, Esther Rincón, Teresa Cejalvo, Irene Izquierdo, Inmaculada Montanuy, Marcos Timón
After many years of promising clinical results splashed with some serious adverse events, gene therapy has finally reached maturity, as demonstrated by the increasing number of medicinal products approved for commercialization by regulatory authorities. The approved products tackle monogenetic inherited diseases as well as cancer, include both in vivo and ex vivo approaches, and comprise mostly gene additions but also a genome-edited product, demonstrating proof of concept for most gene therapy modalities. Uncertainties still remain, especially on their long-term safety and efficacy, which can only be solved with time. These successes should not lead to self-complacency but rather stimulate the development of necessary improvements concerning manufacturing or the safety and efficacy profile. Here, we review the different categories of gene therapy medicinal products and highlight potential areas for improvement. Products approved for commercialization are taken as the basis for the discussion, since information on their assessment is publicly available. New products and manufacturing approaches under development are also reviewed, with an emphasis on the regulatory challenges expected for some of them.
{"title":"The Coming of Age of Gene Therapy for the Treatment of Human Diseases: A Regulatory Perspective.","authors":"Laura Rodríguez, Pablo de Felipe, Raquel Martín, Jorge Martínez, David Ordóñez, Susana Rojo, Juan Fernando Martínez, Esther Rincón, Teresa Cejalvo, Irene Izquierdo, Inmaculada Montanuy, Marcos Timón","doi":"10.1177/10430342251372047","DOIUrl":"10.1177/10430342251372047","url":null,"abstract":"<p><p>After many years of promising clinical results splashed with some serious adverse events, gene therapy has finally reached maturity, as demonstrated by the increasing number of medicinal products approved for commercialization by regulatory authorities. The approved products tackle monogenetic inherited diseases as well as cancer, include both <i>in vivo</i> and <i>ex vivo</i> approaches, and comprise mostly gene additions but also a genome-edited product, demonstrating proof of concept for most gene therapy modalities. Uncertainties still remain, especially on their long-term safety and efficacy, which can only be solved with time. These successes should not lead to self-complacency but rather stimulate the development of necessary improvements concerning manufacturing or the safety and efficacy profile. Here, we review the different categories of gene therapy medicinal products and highlight potential areas for improvement. Products approved for commercialization are taken as the basis for the discussion, since information on their assessment is publicly available. New products and manufacturing approaches under development are also reviewed, with an emphasis on the regulatory challenges expected for some of them.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1103-1117"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144952046","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01Epub Date: 2025-08-29DOI: 10.1177/10430342251372041
Rodrigo A Redondo-Frutos, Pedro Justicia-Lirio, Carmen Barbero-Jimenez, Maider Garnica, Lucia Trudu, Paula Rodriguez-Marquez, Juan J Lasarte, Felipe Prosper, Juan R Rodriguez-Madoz
Chimeric antigen receptor (CAR) T cell therapy has revolutionized treatment for hematological malignancies, yet translating this success to solid tumors remains challenging. Major obstacles include antigen heterogeneity, on-target off-tumor toxicity, limited infiltration and persistence, and the immunosuppressive tumor microenvironment (TME). The present review discusses recent engineering strategies designed to overcome these barriers. Innovations such as affinity-tuned and logic-gated CARs improve specificity and safety, while multi-antigen targeting helps address tumor heterogeneity by avoiding antigen escape. Gene-editing approaches enhance CAR T cell fitness by promoting memory phenotypes, metabolic resilience, and resistance to inhibitory signals imposed by the immunosuppressive TME. Additional modifications improve trafficking, enable extracellular matrix degradation, and reprogram CAR T cells to withstand the hostile conditions of the TME. Together, these advances reflect a growing shift toward rational CAR design and synthetic immunology, with the goal of achieving durable and safe responses in solid tumors. Early clinical trials show promise, and continued translational efforts will be key to unlocking the full therapeutic potential of CAR T cells in this setting.
{"title":"Genetic Engineering in CAR T Cells for Solid Tumors: Current State, Barriers and Future Developments.","authors":"Rodrigo A Redondo-Frutos, Pedro Justicia-Lirio, Carmen Barbero-Jimenez, Maider Garnica, Lucia Trudu, Paula Rodriguez-Marquez, Juan J Lasarte, Felipe Prosper, Juan R Rodriguez-Madoz","doi":"10.1177/10430342251372041","DOIUrl":"10.1177/10430342251372041","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T cell therapy has revolutionized treatment for hematological malignancies, yet translating this success to solid tumors remains challenging. Major obstacles include antigen heterogeneity, on-target off-tumor toxicity, limited infiltration and persistence, and the immunosuppressive tumor microenvironment (TME). The present review discusses recent engineering strategies designed to overcome these barriers. Innovations such as affinity-tuned and logic-gated CARs improve specificity and safety, while multi-antigen targeting helps address tumor heterogeneity by avoiding antigen escape. Gene-editing approaches enhance CAR T cell fitness by promoting memory phenotypes, metabolic resilience, and resistance to inhibitory signals imposed by the immunosuppressive TME. Additional modifications improve trafficking, enable extracellular matrix degradation, and reprogram CAR T cells to withstand the hostile conditions of the TME. Together, these advances reflect a growing shift toward rational CAR design and synthetic immunology, with the goal of achieving durable and safe responses in solid tumors. Early clinical trials show promise, and continued translational efforts will be key to unlocking the full therapeutic potential of CAR T cells in this setting.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1138-1153"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144952018","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01Epub Date: 2025-04-21DOI: 10.1089/hum.2025.006
Melina Weiß, Mareike Selig, Johannes Friedrich, Anna Wierczeiko, Stefan Diederich, Helen Sigel, Janna Bredow, Florian S Eichler, Amanda Nagy, Denise Seyler, Laura Holthöfer, Susanne Gerber, Susann Schweiger, Matthias Linke, Annette Bley
Canavan disease (CD) is a rare autosomal recessive leukodystrophy caused by biallelic pathogenic variants in the ASPA gene. CD is characterized by developmental delay, macrocephaly, and abnormal muscle tone. The biochemical diagnosis is confirmed by increased N-acetylaspartic acid levels. The phenotypic presentation varies, with 85-90% of individuals exhibiting the severe, typical form, while 10-15% present with a milder, atypical form. Here we report on five patients with a clinical and biochemically proven diagnosis in whom a second pathogenic variant had not yet been identified. Targeted long-read sequencing of the entire ASPA gene revealed an SVA_E retrotransposable element located in intron 4 that had been missed by standard short-read-based diagnostic procedures. Haplotype analysis of all patients showed linkage of the SVA_E element with a noncoding variant in intron 1. Functional characterization of the SVA_E element suggests that transcripts of the affected allele are prone to highly efficient mRNA degradation processes. These findings enhance the precision of genetic diagnostics and enable improved guidance for families as well as facilitating potential access to targeted therapies.
{"title":"Deep Intronic SVA_E Retrotransposition as a Novel Factor in Canavan Disease Pathogenesis.","authors":"Melina Weiß, Mareike Selig, Johannes Friedrich, Anna Wierczeiko, Stefan Diederich, Helen Sigel, Janna Bredow, Florian S Eichler, Amanda Nagy, Denise Seyler, Laura Holthöfer, Susanne Gerber, Susann Schweiger, Matthias Linke, Annette Bley","doi":"10.1089/hum.2025.006","DOIUrl":"10.1089/hum.2025.006","url":null,"abstract":"<p><p>Canavan disease (CD) is a rare autosomal recessive leukodystrophy caused by biallelic pathogenic variants in the <i>ASPA</i> gene. CD is characterized by developmental delay, macrocephaly, and abnormal muscle tone. The biochemical diagnosis is confirmed by increased <i>N</i>-acetylaspartic acid levels. The phenotypic presentation varies, with 85-90% of individuals exhibiting the severe, typical form, while 10-15% present with a milder, atypical form. Here we report on five patients with a clinical and biochemically proven diagnosis in whom a second pathogenic variant had not yet been identified. Targeted long-read sequencing of the entire <i>ASPA</i> gene revealed an SVA_E retrotransposable element located in intron 4 that had been missed by standard short-read-based diagnostic procedures. Haplotype analysis of all patients showed linkage of the SVA_E element with a noncoding variant in intron 1. Functional characterization of the SVA_E element suggests that transcripts of the affected allele are prone to highly efficient mRNA degradation processes. These findings enhance the precision of genetic diagnostics and enable improved guidance for families as well as facilitating potential access to targeted therapies.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1248-1256"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12596875/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144008840","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01Epub Date: 2025-04-22DOI: 10.1089/hum.2024.265
Sofia Fernandes, Júlia Guerra, Mariana V Ferreira, Ana Sofia Coroadinha
Over the last two decades, adeno-associated viruses (AAVs) have been widely used as viral vectors in gene therapy due to their ability to infect both dividing and nondividing cells, broad tissue specificity, and favorable safety profile. Recombinant AAV (rAAV) production requires a helper virus, typically adenovirus (AdV), which provides essential genes for AAV replication. However, the increasing demand for safer and more efficient rAAV production methods led to the need to develop helper plasmids with minimal AdV components. In this study, we evaluate the impact of AdV E2 and E4 in the productivity and genome packaging of rAAV serotypes 2, 5, 8, and 9, produced by transient transfection. We designed and tested eight novel helper plasmids with different deletions in E2 and E4 genes. Results indicated that deletions in these genes significantly affected rAAV productivity and packaging, particularly for serotypes 8 and 9. Helper plasmids containing minimal essential genes-E2-DBP, E4orf6, and VA RNA-showed near to 10-fold reduction in viral genome packaging compared to the control. However, including E2 L4-22/33K and E4orf3 regions significantly improved viral production, particularly for serotypes 8, and 9. In this study, we also demonstrated that the full E4 gene is crucial to achieving high full-empty ratios, minimizing the production of empty capsids, and enhancing viral release into the culture medium of rAAV8. Accordingly, we created a smaller plasmid, without adenoviral structural proteins that allows a similar rAAV production across all tested serotypes. Overall, these findings provide insights into the genetic requirements for efficient rAAV production and highlight the importance of the E2 and E4 regions for optimizing viral yield and quality. This approach could lead to the development of improved strategies for large-scale rAAV vector production by enabling safer and more cost-effective systems.
{"title":"Deciphering Key Adenoviral Elements in the Production of Recombinant Adeno-Associated Virus Vectors.","authors":"Sofia Fernandes, Júlia Guerra, Mariana V Ferreira, Ana Sofia Coroadinha","doi":"10.1089/hum.2024.265","DOIUrl":"10.1089/hum.2024.265","url":null,"abstract":"<p><p>Over the last two decades, adeno-associated viruses (AAVs) have been widely used as viral vectors in gene therapy due to their ability to infect both dividing and nondividing cells, broad tissue specificity, and favorable safety profile. Recombinant AAV (rAAV) production requires a helper virus, typically adenovirus (AdV), which provides essential genes for AAV replication. However, the increasing demand for safer and more efficient rAAV production methods led to the need to develop helper plasmids with minimal AdV components. In this study, we evaluate the impact of AdV E2 and E4 in the productivity and genome packaging of rAAV serotypes 2, 5, 8, and 9, produced by transient transfection. We designed and tested eight novel helper plasmids with different deletions in E2 and E4 genes. Results indicated that deletions in these genes significantly affected rAAV productivity and packaging, particularly for serotypes 8 and 9. Helper plasmids containing minimal essential genes-E2-DBP, E4orf6, and VA RNA-showed near to 10-fold reduction in viral genome packaging compared to the control. However, including E2 L4-22/33K and E4orf3 regions significantly improved viral production, particularly for serotypes 8, and 9. In this study, we also demonstrated that the full E4 gene is crucial to achieving high full-empty ratios, minimizing the production of empty capsids, and enhancing viral release into the culture medium of rAAV8. Accordingly, we created a smaller plasmid, without adenoviral structural proteins that allows a similar rAAV production across all tested serotypes. Overall, these findings provide insights into the genetic requirements for efficient rAAV production and highlight the importance of the E2 and E4 regions for optimizing viral yield and quality. This approach could lead to the development of improved strategies for large-scale rAAV vector production by enabling safer and more cost-effective systems.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1199-1210"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143981324","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01Epub Date: 2025-06-04DOI: 10.1089/hum.2024.256
Jessica Trundle, Alexis Boulinguiez, Ngoc Lu-Nguyen, James March, Alberto Malerba, Linda Popplewell
Duchenne muscular dystrophy (DMD) is a severe, progressive genetic disorder primarily affecting boys, characterized by muscle degeneration due to mutations in the DMD gene encoding dystrophin, a crucial protein for muscle fiber integrity. The disease leads to significant muscle weakness and eventually to loss of ambulation. Adeno-associated viral (AAV)-microdystrophin (MD) gene therapy shows promise in preclinical and clinical settings. However, muscle fibrosis, a consequence of chronic inflammation and extracellular matrix remodeling, exacerbates disease progression and may hinder therapeutic efficacy. Periostin, a matricellular protein involved in fibrosis, is upregulated in DMD rodent models and correlates with collagen deposition. We previously developed an antisense oligonucleotide strategy to induce exon 17 skipping and so reduce periostin expression and collagen accumulation in the fibrotic D2.mdx mouse model of DMD. Here, we investigated the combined effects of periostin modulation and AAV-MD1 treatment. We found that systemic periostin splicing modulation significantly improved muscle function, assessed by forelimb grip strength and treadmill performance. Importantly, periostin exon skipping increased the MD protein expression. These findings suggest that targeting periostin in conjunction with MD therapy could represent a valid therapeutic strategy for DMD.
{"title":"Periostin Exon 17 Skipping Enhances the Efficacy of Local Adeno-Associated Viral-Microdystrophin Administration in a Fibrotic Model of Duchenne Muscular Dystrophy.","authors":"Jessica Trundle, Alexis Boulinguiez, Ngoc Lu-Nguyen, James March, Alberto Malerba, Linda Popplewell","doi":"10.1089/hum.2024.256","DOIUrl":"10.1089/hum.2024.256","url":null,"abstract":"<p><p>Duchenne muscular dystrophy (DMD) is a severe, progressive genetic disorder primarily affecting boys, characterized by muscle degeneration due to mutations in the DMD gene encoding dystrophin, a crucial protein for muscle fiber integrity. The disease leads to significant muscle weakness and eventually to loss of ambulation. Adeno-associated viral (AAV)-microdystrophin (MD) gene therapy shows promise in preclinical and clinical settings. However, muscle fibrosis, a consequence of chronic inflammation and extracellular matrix remodeling, exacerbates disease progression and may hinder therapeutic efficacy. Periostin, a matricellular protein involved in fibrosis, is upregulated in DMD rodent models and correlates with collagen deposition. We previously developed an antisense oligonucleotide strategy to induce exon 17 skipping and so reduce periostin expression and collagen accumulation in the fibrotic D2.<i>mdx</i> mouse model of DMD. Here, we investigated the combined effects of periostin modulation and AAV-MD1 treatment. We found that systemic periostin splicing modulation significantly improved muscle function, assessed by forelimb grip strength and treadmill performance. Importantly, periostin exon skipping increased the MD protein expression. These findings suggest that targeting periostin in conjunction with MD therapy could represent a valid therapeutic strategy for DMD.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1257-1267"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144215695","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01Epub Date: 2025-08-21DOI: 10.1177/10430342251372063
Joaquin Delgadillo, Francisco Martin, Gloria Carmona, Josep M Canals, Paula Rio, Cristina Eguizabal, Felipe Prosper, Cristina Avendaño-Solá
Gene therapy has revolutionized modern medicine by offering innovative treatments for genetic disorders, cancers, and immune-related conditions through technologies such as viral vector delivery, genome editing, and genetically modified cell therapies. Despite significant advancements, the classification of gene therapy medicinal products (GTMPs) as genetically modified organisms (GMOs) under EU legislation imposes significant regulatory burdens, hindering early and timely patient access to such therapies. Current GMO regulations, originally designed for agricultural biotechnology, require environmental risk assessments (ERAs) and additional approvals, creating delays and increasing costs-with a particularly negative impact on early academic research. This article examines the scientific and regulatory discrepancies in classifying GTMPs as GMOs, arguing that replication-deficient vectors and non-persistent modified cells may not meet the criteria for GMOs. We highlight the negative impact of GMO requirements on clinical trial feasibility in Europe compared to the U.S., where a categorical exclusion from ERA applies to investigational medicinal products. Proposed solutions include adopting a risk-based regulatory model, harmonizing ERA processes under the revised EU Clinical Trials Regulation, and establishing exemptions for low-risk therapies. By aligning regulatory frameworks with scientific evidence, policymakers can accelerate the translation of gene therapies while maintaining safety standards, ultimately improving patient access to these transformative treatments.
{"title":"Regulatory Strategies for Accelerating the Translation of Gene Therapies to Clinical Practice: Focus on GMO Considerations.","authors":"Joaquin Delgadillo, Francisco Martin, Gloria Carmona, Josep M Canals, Paula Rio, Cristina Eguizabal, Felipe Prosper, Cristina Avendaño-Solá","doi":"10.1177/10430342251372063","DOIUrl":"10.1177/10430342251372063","url":null,"abstract":"<p><p>Gene therapy has revolutionized modern medicine by offering innovative treatments for genetic disorders, cancers, and immune-related conditions through technologies such as viral vector delivery, genome editing, and genetically modified cell therapies. Despite significant advancements, the classification of gene therapy medicinal products (GTMPs) as genetically modified organisms (GMOs) under EU legislation imposes significant regulatory burdens, hindering early and timely patient access to such therapies. Current GMO regulations, originally designed for agricultural biotechnology, require environmental risk assessments (ERAs) and additional approvals, creating delays and increasing costs-with a particularly negative impact on early academic research. This article examines the scientific and regulatory discrepancies in classifying GTMPs as GMOs, arguing that replication-deficient vectors and non-persistent modified cells may not meet the criteria for GMOs. We highlight the negative impact of GMO requirements on clinical trial feasibility in Europe compared to the U.S., where a categorical exclusion from ERA applies to investigational medicinal products. Proposed solutions include adopting a risk-based regulatory model, harmonizing ERA processes under the revised EU Clinical Trials Regulation, and establishing exemptions for low-risk therapies. By aligning regulatory frameworks with scientific evidence, policymakers can accelerate the translation of gene therapies while maintaining safety standards, ultimately improving patient access to these transformative treatments.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1154-1158"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144951996","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01Epub Date: 2025-08-29DOI: 10.1177/10430342251372091
Daniel Palacios-Alonso, Miriam Bazan-Peregrino, Marta Zalacain, Lucía Marrodán, Ana Mato-Berciano, Jaime Gállego Pérez-Larraya, Iker Ausejo-Mauleon, Ana Patiño-García, Lilian Zavala-Romero, Manel Cascalló, Ramon Alemany, Marisol González-Huarriz, Marta M Alonso
Among solid pediatric tumors, brain tumors are the leading cause of cancer-related mortality. While survival rates have improved for certain pediatric brain tumor subtypes, the overall prognosis remains poor. Consequently, there is an urgent need for novel therapies that are not only effective but also less toxic. Oncolytic viruses have emerged as promising therapeutic agents due to their ability to selectively replicate in tumor cells while sparing healthy tissue and their potential to induce systemic antitumor immune responses. VCN-01 is a replication-competent oncolytic adenovirus whose efficacy has been demonstrated in clinical trials after systemic administration in combination with chemotherapy. Evidence of antitumor activity has also been obtained after intracranial administration in preclinical models of various brain tumors, including high-grade gliomas. However, before progressing to clinical trials for those indications, it is essential to assess the safety of its intracranial administration. In this study, we evaluated the toxicity and biodistribution of VCN-01 following intracranial injection in a Syrian hamster model. Two viral doses were tested: 1.5 × 109 and 1.5 × 1010 viral particles (vp)/animal, corresponding to 5 and 50 times the starting clinical dose (1010 vp/patient), respectively. Our toxicity analysis revealed a favorable safety profile, with no adverse effects observed following administration. Biodistribution studies demonstrated that VCN-01 primarily remained confined to the brain, with only minimal presence detected in peripheral tissues. The neutralizing antibody response against the virus was stronger in females than in males, correlating with a lower detection of vp in females compared with males. In conclusion, these findings support the safety of intracranial administration of VCN-01 and provide a strong rationale for its further development as a therapeutic option for patients with brain tumors.
{"title":"Toxicity and Biodistribution of the Oncolytic Virus VCN-01 Following Intracranial Injection in Syrian Hamsters.","authors":"Daniel Palacios-Alonso, Miriam Bazan-Peregrino, Marta Zalacain, Lucía Marrodán, Ana Mato-Berciano, Jaime Gállego Pérez-Larraya, Iker Ausejo-Mauleon, Ana Patiño-García, Lilian Zavala-Romero, Manel Cascalló, Ramon Alemany, Marisol González-Huarriz, Marta M Alonso","doi":"10.1177/10430342251372091","DOIUrl":"10.1177/10430342251372091","url":null,"abstract":"<p><p>Among solid pediatric tumors, brain tumors are the leading cause of cancer-related mortality. While survival rates have improved for certain pediatric brain tumor subtypes, the overall prognosis remains poor. Consequently, there is an urgent need for novel therapies that are not only effective but also less toxic. Oncolytic viruses have emerged as promising therapeutic agents due to their ability to selectively replicate in tumor cells while sparing healthy tissue and their potential to induce systemic antitumor immune responses. VCN-01 is a replication-competent oncolytic adenovirus whose efficacy has been demonstrated in clinical trials after systemic administration in combination with chemotherapy. Evidence of antitumor activity has also been obtained after intracranial administration in preclinical models of various brain tumors, including high-grade gliomas. However, before progressing to clinical trials for those indications, it is essential to assess the safety of its intracranial administration. In this study, we evaluated the toxicity and biodistribution of VCN-01 following intracranial injection in a Syrian hamster model. Two viral doses were tested: 1.5 × 10<sup>9</sup> and 1.5 × 10<sup>10</sup> viral particles (vp)/animal, corresponding to 5 and 50 times the starting clinical dose (10<sup>10</sup> vp/patient), respectively. Our toxicity analysis revealed a favorable safety profile, with no adverse effects observed following administration. Biodistribution studies demonstrated that VCN-01 primarily remained confined to the brain, with only minimal presence detected in peripheral tissues. The neutralizing antibody response against the virus was stronger in females than in males, correlating with a lower detection of vp in females compared with males. In conclusion, these findings support the safety of intracranial administration of VCN-01 and provide a strong rationale for its further development as a therapeutic option for patients with brain tumors.</p>","PeriodicalId":13007,"journal":{"name":"Human gene therapy","volume":" ","pages":"1237-1247"},"PeriodicalIF":4.0,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144952050","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}