1
|
Kumar ARK, Low J, Lim J, Myint B, Sun X, Wu L, Cheng HS, Yip S, Ming Cheng CZ, Manoharan T, Quek YJ, Shou Y, Tian JS, Ng YY, Gascoigne NRJ, Tan NS, Sugimura R, Chia G, Sze Cheung AM, Yawata M, Tay A. Non-viral, high throughput genetic engineering of primary immune cells using nanostraw-mediated transfection. Biomaterials 2025; 317:123079. [PMID: 39842078 DOI: 10.1016/j.biomaterials.2024.123079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Transfection of proteins, mRNA, and chimeric antigen receptor (CAR) transgenes into immune cells remains a critical bottleneck in cell manufacturing. Current methods, such as viruses and bulk electroporation, are hampered by low transfection efficiency, unintended transgene integration, and significant cell perturbation. The Nanostraw Electro-actuated Transfection (NExT) technology offers a solution by using high aspect-ratio nanostraws and localized electric fields to precisely deliver biomolecules into cells with minimal disruption. We demonstrate that NExT can deliver proteins, polysaccharides, and mRNA into primary human CD8+ and CD4+ T cells, and achieve CRISPR/Cas9 gene knockout of CXCR4 and TRAC in CD8+ T cells. We showcase NExT's versatility across a range of primary human immune cells, including CD4+ T cells, γδ-T cells, dendritic cells, NK cells, Treg cells, macrophages, and neutrophils. Finally, we developed a scalable, high-throughput multiwell NExT system capable of transfecting over 14 million cells and delivering diverse cargoes into multiple cell types from various donors simultaneously. This technology holds promise for streamlining high-throughput screening of allogeneic donors and reducing optimization costs for large-scale CAR-immune cell transfection.
Collapse
Affiliation(s)
- Arun R K Kumar
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Jessalyn Low
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Jet Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Ba Myint
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Xinhong Sun
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Ling Wu
- Immunology Translational Research Programme and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117545, Singapore
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Sophronia Yip
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China; Centre for Translational Stem Cell Biology, Hong Kong SAR, China
| | - Cyrus Zai Ming Cheng
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Thamizhanban Manoharan
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Ying Jie Quek
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Johann Shane Tian
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Yu Yang Ng
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117545, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Rio Sugimura
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China; Centre for Translational Stem Cell Biology, Hong Kong SAR, China
| | - Gloryn Chia
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Alice Man Sze Cheung
- Department of Haematology, Singapore General Hospital, Singapore, 169608, Singapore; SingHealth Duke-NUS Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, 168753, Singapore
| | - Makoto Yawata
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 119077, Singapore
| | - Andy Tay
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore; NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore.
| |
Collapse
|
2
|
Wang D, Stevens G, Flotte TR. Gene therapy then and now: A look back at changes in the field over the past 25 years. Mol Ther 2025; 33:1889-1902. [PMID: 40022444 DOI: 10.1016/j.ymthe.2025.02.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025] Open
Abstract
Since the inception of Molecular Therapy in 2000, the field of gene therapy has made remarkable progress, evolving from no approved clinical products to 23 clinical gene therapy products today. In this review, we aim to capture the transformative changes in the field by surveying the literature over this period, with a particular focus on advancements in gene delivery vector technology, disease and tissue targeting, and the revolutionary molecular tools that have become central to the field. We also discuss the current challenges facing gene therapy and the need for greater collaboration to ensure its accessibility worldwide.
Collapse
Affiliation(s)
- Dan Wang
- Department of Genetic and Cellular Medicine, UMass Chan Medical School, Worcester, MA, USA
| | - Gregg Stevens
- Lamar Soutter Library, UMass Chan Medical School, Worcester, MA, USA
| | - Terence R Flotte
- Department of Genetic and Cellular Medicine, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
3
|
Staal FJT, Pike-Overzet K, de Kivit S, Ott de Bruin L, Mamede L, Pergent M, Prevot J, Rothe M, Schambach A, Lankester A. Safety and efficacy of gene therapy for RAG1-deficient SCID. Mol Ther 2025; 33:1869-1870. [PMID: 40107272 DOI: 10.1016/j.ymthe.2025.02.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
| | | | | | | | - Lucia Mamede
- International Patient Organization for Primary Immune Deficiencies (IPOPI), Brussels, Belgium
| | - Martine Pergent
- International Patient Organization for Primary Immune Deficiencies (IPOPI), Brussels, Belgium
| | - Johan Prevot
- International Patient Organization for Primary Immune Deficiencies (IPOPI), Brussels, Belgium
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
4
|
Williams DA, Kohn DB, Thrasher AJ. Ex vivo modification of hematopoietic stem and progenitor cells for gene therapy. Mol Ther 2025; 33:2141-2153. [PMID: 40176348 DOI: 10.1016/j.ymthe.2025.03.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025] Open
Abstract
The development of viral vectors has been particularly critical for genetic therapies of hematological diseases. Before the development of retrovirus vectors (RVVs), gene transfer into mammalian cells was accomplished by transduction of DNA plasmids by chemical means and later by electroporation. The main limitation of these methods is the inefficiency of transfer of intact sequences, and particularly with electroporation significant cell death of the manipulated cells. The earliest successful human gene therapy trials utilized γ-RVVs and many of the techniques developed in the 1980s. A breakthrough for the field was the exploitation and development of HIV for transfer vectors, termed lentivirus vectors. In this review, we highlight uses of retro- and lentivirus vectors in monogenic diseases in which hematopoietic stem cells are used in the autologous setting to treat immunodeficiencies, hemoglobinopathies and metabolic diseases. The three authors' perspective represent experiences in the field over four decades that encompasses both basic translational research and development and oversight of early and ongoing gene therapy trials utilizing viral vectors.
Collapse
Affiliation(s)
- David A Williams
- Boston Children's Hospital, Dana-Farber & Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115 USA.
| | - Donald B Kohn
- Department of Pediatrics (Hematology/Oncology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Adrian J Thrasher
- Molecular and Cellular Immunology, GOS Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
5
|
Lenders M, Menke ER, Brand E. Progress and Challenges in the Treatment of Fabry Disease. BioDrugs 2025:10.1007/s40259-025-00723-3. [PMID: 40310476 DOI: 10.1007/s40259-025-00723-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2025] [Indexed: 05/02/2025]
Abstract
Fabry disease is a rare but life-threatening, X-linked, inherited lysosomal storage disorder in which globotriaosylceramide is insufficiently metabolized because of reduced α-galactosidase A activity. Cellular globotriaosylceramide accumulation causes a multisystemic disease, which, if left untreated, reduces life expectancy in female and male individuals by around 10 and 20 years, respectively, leading to progressive renal failure, hypertrophic cardiomyopathy, cardiac arrhythmia, and premature cerebral infarction. The method of choice for confirming the diagnosis is the determination of reduced α-galactosidase A activity in leukocytes in male individuals and the molecular genetic detection of a disease-causing mutation in female individuals. Current approved treatment includes enzyme replacement therapy (agalsidase alfa [0.2 mg/kg body weight], agalsidase beta or pegunigalsidase alfa [both 1.0 mg/kg body weight]) every other week intravenously or, if a responding ('amenable') α-galactosidase A mutation is present, oral pharmacological chaperone therapy (migalastat 123 mg, every other day). Future therapeutic options may include substrate reduction therapy, gene therapy, messenger RNA therapy, and/or vesicle-packaged enzyme replacement therapy. This review presents current and future treatment options with advantages and disadvantages of the different treatment options.
Collapse
Affiliation(s)
- Malte Lenders
- Internal Medicine D (Nephrology, Hypertension and Rheumatology), and Interdisciplinary Fabry Center (IFAZ), University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| | - Elise Raphaela Menke
- Internal Medicine D (Nephrology, Hypertension and Rheumatology), and Interdisciplinary Fabry Center (IFAZ), University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| | - Eva Brand
- Internal Medicine D (Nephrology, Hypertension and Rheumatology), and Interdisciplinary Fabry Center (IFAZ), University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany.
| |
Collapse
|
6
|
Ay C, Reinisch A. Gene therapy: principles, challenges and use in clinical practice. Wien Klin Wochenschr 2025; 137:261-271. [PMID: 38713227 PMCID: PMC12081535 DOI: 10.1007/s00508-024-02368-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/12/2024] [Indexed: 05/08/2024]
Abstract
INTRODUCTION Gene therapy is an emerging topic in medicine. The first products have already been licensed in the European Union for the treatment of immune deficiency, spinal muscular atrophy, hemophilia, retinal dystrophy, a rare neurotransmitter disorder and some hematological cancers, while many more are being assessed in preclinical and clinical trials. OBJECTIVE The purpose of this review is to provide an overview of the core principles of gene therapy along with information on challenges and risks. Benefits, adverse effects and potential risks are illustrated based on the examples of hemophilia and spinal muscular atrophy. RESULTS At present, in-vitro and in-vivo gene addition or gene augmentation is the most commonly established type of gene therapy. More recently, more sophisticated and precise approaches such as in situ gene editing have moved into focus. However, all types of gene therapy require long-term observation of treated patients to ensure safety, efficacy, predictability and durability. Important safety concerns include immune reactions to the vector, the foreign DNA or the new protein resulting from gene therapy, and a remaining low cancer risk based on insertional mutagenesis. Ethical and regulatory issues need to be addressed, and new reimbursement models are called for to ease the financial burden that this new treatment poses for the health care system. CONCLUSION Gene therapy holds great promise for considerable improvement or even cure of genetic diseases with serious clinical consequences. However, a number of questions and issues need to be clarified to ensure broad accessibility of safe and efficacious products.
Collapse
Affiliation(s)
- Cihan Ay
- Department of Medicine I, Clinical Division of Haematology and Haemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Andreas Reinisch
- Department of Medicine, Division of Hematology & Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Auenbruggerplatz 38, 8036, Graz, Austria.
| |
Collapse
|
7
|
Ramos-Hernández I, Fuster-García C, Aguilar-González A, Lozano-Vinagre M, Guenechea-Amurrio G, Sanchez-Luque F, Gonçalves MFV, Cathomen T, Muñoz P, Molina-Estévez F, Martín F. Donor insertion into CX3CR1 allows epigenetic modulation of a constitutive promoter on hematopoietic stem cells and its activation upon myeloid differentiation. Nucleic Acids Res 2025; 53:gkaf344. [PMID: 40298109 PMCID: PMC12038399 DOI: 10.1093/nar/gkaf344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025] Open
Abstract
To improve ex vivo gene therapy strategies involving hematopoietic stem and progenitor cells (HSPCs), we propose a novel knock-in strategy (named KI-Ep) aiming to achieve transgene regulation of the inserted cassette through the acquisition of naturally occurring epigenetic marks. Based on this hypothesis, we selected CX3CR1 (a myeloid-specific gene presenting a poised histone signature on primitive HSPCs) as safe harbor to generate KI-Ep HSPCs. We demonstrated that, unlike the expression pattern achieved with lentiviral vectors (LVs), the insertion of a constitutive expression cassette into the intron 1 of the CX3CR1 locus (CX3CR1-I) in HSPCs resulted in very low expression levels in the more primitive HSPCs but, crucially, strong expression in HSPC-differentiated populations (especially myeloid cells), both in vitro and in vivo. Furthermore, we showed that the promoter of the expression cassette inserted into CX3CR1-I acquired epigenetic marks associated with poised genes during the HSPC stage. These marks transitioned to activated histone states upon KI-Ep HSPCs differentiation. In summary, here, we introduce the KI-Ep concept which enables the epigenetic modulation of the inserted transgene during the HSPCs stem cell stages and its subsequent activation upon differentiation.
Collapse
Affiliation(s)
- Iris Ramos-Hernández
- GENYO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Andalusian Regional Government. PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain
- Fundación Pública Andaluza para la Investigación Biosanitaria en Andalucía Oriental Alejandro Otero (FIBAO), Avenida de Madrid, 15, Beiro, 18012 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
| | - Carla Fuster-García
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center—University of Freiburg, 79106 Freiburg, Germany
| | - Araceli Aguilar-González
- GENYO, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research, Andalusian Regional Government. PTS Granada, Avenida de la Ilustración 114, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, School of Pharmacy, University of Granada, Campus Cartuja s/n, 18071 Granada, Spain
| | - María L Lozano-Vinagre
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Guillermo Guenechea-Amurrio
- Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Francisco J Sanchez-Luque
- Institute of Parasitology and Biomedicine ‘López-Neyra’ (Spanish National Research Council), Avda. del Conocimiento 17 (PTS Granada), 18016 Armilla (Granada), Spain
| | - Manuel A F V Gonçalves
- Leiden University Medical Center, Department of Cell and Chemical Biology, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg,79110 Freiburg, Germany
| | - Pilar Muñoz
- Fundación Pública Andaluza para la Investigación Biosanitaria en Andalucía Oriental Alejandro Otero (FIBAO), Avenida de Madrid, 15, Beiro, 18012 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
- Departmento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Campus Fuentenueva, 18071 Granada, Spain
| | - Francisco J Molina-Estévez
- Fundación Pública Andaluza para la Investigación Biosanitaria en Andalucía Oriental Alejandro Otero (FIBAO), Avenida de Madrid, 15, Beiro, 18012 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
- Departmento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Campus Fuentenueva, 18071 Granada, Spain
| | - Francisco Martín
- Fundación Pública Andaluza para la Investigación Biosanitaria en Andalucía Oriental Alejandro Otero (FIBAO), Avenida de Madrid, 15, Beiro, 18012 Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18012 Granada, Spain
- Departamento de Bioquímica y Biología Molecular 3 e Inmunología, Facultad de Medicina, Universidad de Granada, Avda. de la Investigación 11, 18071 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016, 34 Granada, Spain
| |
Collapse
|
8
|
Chang J, Yang X, Zhang T, Sun H, Cheng H, Jia Z, Li Y, Ma S, Sun T, Cao J. High-Throughput Screening to Identify Novel Compounds Affecting the Genome Editing Efficiency of CRISPR System. Molecules 2025; 30:1811. [PMID: 40333840 PMCID: PMC12029788 DOI: 10.3390/molecules30081811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 05/09/2025] Open
Abstract
Genome editing is a promising therapeutic strategy for genetic disorders by modifying the genome precisely, especially the CRISPR/Cas9 system. However, a major limitation of CRISPR/Cas9 in gene therapy is the biosafety issues caused by off-target effects. Compounds that can modulate the genome editing efficiency of the CRISPR/Cas9 system, especially those reducing the off-target effects, are potentially useful pharmacological tools for improving the effectiveness and safety of genome editing. Here, we performed high-throughput screening in HEK 293FT cells to discover compounds that decrease or increase the genome editing efficiency of the CRISPR/Cas9 system from 9930 compounds. After two rounds of screening, we identified that CP-724714, a ErbB2 (HER2) tyrosine kinase inhibitor, decreased the CRISPR/Cas9 efficiency and reduced the off-target effects by suppressing the efficiency of CRISPR/Cas9, and was thus named a CRISPR decelerator (or inhibitor), while Clofarabine, a DNA synthesis inhibitor, increased the efficiency of CRISPR/Cas9, and was named a CRISPR accelerator. We further identified four compounds (Tranilast, Cerulenin, Rosolic acid and Resveratrol) that affected the efficiency of single-strand annealing (SSA) repair. Among them, Tranilast, Cerulenin and Rosolic acid are potential SSA decelerators, while Resveratrol is a potential SSA accelerator. These identified compounds may be useful in optimizing mammalian genetic manipulation techniques.
Collapse
Affiliation(s)
- Jiasong Chang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, China; (J.C.); (X.Y.); (Z.J.); (Y.L.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Xiulong Yang
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, China; (J.C.); (X.Y.); (Z.J.); (Y.L.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Tong Zhang
- Biological Science Research Center, Southwest University, Chongqing 400715, China; (T.Z.); (H.S.); (S.M.)
| | - Hao Sun
- Biological Science Research Center, Southwest University, Chongqing 400715, China; (T.Z.); (H.S.); (S.M.)
| | - Hongying Cheng
- Department of Preschool Education, Lvliang Teachers College, Lvliang 033001, China;
| | - Zhangrong Jia
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, China; (J.C.); (X.Y.); (Z.J.); (Y.L.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Yiying Li
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, China; (J.C.); (X.Y.); (Z.J.); (Y.L.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Sanyuan Ma
- Biological Science Research Center, Southwest University, Chongqing 400715, China; (T.Z.); (H.S.); (S.M.)
| | - Teng Sun
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, China; (J.C.); (X.Y.); (Z.J.); (Y.L.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, China; (J.C.); (X.Y.); (Z.J.); (Y.L.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
9
|
Ottaviano G, Qasim W. Current landscape of vector safety and genotoxicity after hematopoietic stem or immune cell gene therapy. Leukemia 2025:10.1038/s41375-025-02585-8. [PMID: 40200078 DOI: 10.1038/s41375-025-02585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Malignant transformation of gene modified haematopoietic stem cells caused anxiety following adverse events in early clinical trials using gamma-retroviral vectors (γRV) to correct haematopoietic stem cells (HSC) in monogenic immune disorders. Adoption of HIV-derived lentiviral vectors (LV) with SIN (self-inactivating) configurations greatly reduced risks and subsequently hundreds of patients have been dosed with HSC gene therapy for blood, immune and metabolic conditions. Nevertheless, as experience builds, it's now well recognised that vector integration can drive clonal expansions and these may carry long term safety risks. Documented cases of haematological malignancy after SIN-LV gene therapy have recently emerged, in particular where heterologous retroviral promoters were employed and there are concerns around certain insulator elements and other possible contributors to clonal expansions. Similarly, tens of thousands of subjects have now received engineered T cell products, and longstanding dogma that mature T cells cannot be transformed is being questioned, with reports of a small number of malignant transformation events and wider concerns around secondary malignancies in some groups of patients. We summarize current clinical information and revisit genotoxicity risks following ex-vivo gene modification of HSC and T cells.
Collapse
Affiliation(s)
- Giorgio Ottaviano
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
- Molecular and Cellular Immunology, University College London, London, UK.
| | - Waseem Qasim
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Molecular and Cellular Immunology, University College London, London, UK
| |
Collapse
|
10
|
Assessing the risks of engineered T cells. Nat Med 2025; 31:1079-1080. [PMID: 40065174 DOI: 10.1038/s41591-025-03598-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
|
11
|
Jadlowsky JK, Hexner EO, Marshall A, Grupp SA, Frey NV, Riley JL, Veloso E, McConville H, Rogal W, Czuczman C, Hwang WT, Li Y, Leskowitz RM, Farrelly O, Karar J, Christensen S, Barber-Rotenberg J, Gaymon A, Aronson N, Bernstein W, Melenhorst JJ, Roche AM, Everett JK, Zolnoski SA, McFarland AG, Reddy S, Petrichenko A, Cook EJ, Lee C, Gonzalez VE, Alexander K, Kulikovskaya I, Ramírez-Fernández Á, Minehart JC, Ruella M, Gill SI, Schuster SJ, Cohen AD, Garfall AL, Shah PD, Porter DL, Maude SL, Levine BL, Siegel DL, Chew A, McKenna S, Lledo L, Davis MM, Plesa G, Herbst F, Stadtmauer EA, Tebas P, DiNofia A, Haas A, Haas NB, Myers R, O'Rourke DM, Svoboda J, Tanyi JL, Aplenc R, Jacobson JM, Ko AH, Cohen RB, June CH, Bushman FD, Fraietta JA. Long-term safety of lentiviral or gammaretroviral gene-modified T cell therapies. Nat Med 2025; 31:1134-1144. [PMID: 39833408 DOI: 10.1038/s41591-024-03478-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Long-term risks of gene therapy are not fully understood. In this study, we evaluated safety outcomes in 783 patients over more than 2,200 total patient-years of observation from 38 T cell therapy trials. The trials employed integrating gammaretroviral or lentiviral vectors to deliver engineered receptors to target HIV-1 infection or cancer. Eighteen patients (2.3%) developed secondary malignancies after treatment, with a median onset of 1.94 years (range: 51 d to 14 years). Where possible, incident tumor samples were analyzed for vector copy number, revealing no evidence of high-level marking or other indications of insertional mutagenesis. One T cell lymphoma was detected, but malignant T cells were not marked by vector integration. Analysis of vector integration sites in 176 patients revealed no pathological insertions linked to secondary malignancies, although, in some cases, integration in or near specific genes, including tumor suppressor genes, was associated with modest clonal expansion and sustained T cell persistence. These findings highlight the safety of engineered T cell therapies.
Collapse
Affiliation(s)
- Julie K Jadlowsky
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth O Hexner
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Marshall
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephan A Grupp
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Noelle V Frey
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James L Riley
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Veloso
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Holly McConville
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter Rogal
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cory Czuczman
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei-Ting Hwang
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yimei Li
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel M Leskowitz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olivia Farrelly
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayashree Karar
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon Christensen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie Barber-Rotenberg
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Avery Gaymon
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi Aronson
- Department of Medicine, Division of Infectious Diseases, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Wendy Bernstein
- Department of Medicine, Division of Infectious Diseases, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jan Joseph Melenhorst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aoife M Roche
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John K Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sonja A Zolnoski
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander G McFarland
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shantan Reddy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angelina Petrichenko
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma J Cook
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carole Lee
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vanessa E Gonzalez
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen Alexander
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Irina Kulikovskaya
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ángel Ramírez-Fernández
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janna C Minehart
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Schuster
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam D Cohen
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alfred L Garfall
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Payal D Shah
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David L Porter
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon L Maude
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Donald L Siegel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anne Chew
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen McKenna
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lester Lledo
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan M Davis
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Friederike Herbst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Stadtmauer
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pablo Tebas
- Department of Medicine, Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda DiNofia
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew Haas
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Naomi B Haas
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Regina Myers
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Donald M O'Rourke
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jakub Svoboda
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janos L Tanyi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Aplenc
- Department of Pediatrics, Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey M Jacobson
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew H Ko
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Roger B Cohen
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Rist M, Kaku M, Coffin JM. Ex vivo HIV DNA integration in STAT3 drives T cell persistence-A model of HIV-associated T cell lymphoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646272. [PMID: 40236153 PMCID: PMC11996357 DOI: 10.1101/2025.03.31.646272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Oncogenic retroviruses are known for their pathogenesis via insertional mutagenesis, in which the presence of a provirus and its transcriptional control elements alter the expression of a nearby or surrounding host gene. There are reports of proviral integration driving oncogenesis in people with HIV and the use of HIV-derived vectors for gene therapy has raised concern about oncogenic side effects. To study this issue, we used an ex vivo human CD4+ T cell infection model developed in our laboratory to identify HIV-1 integration sites that might influence cell proliferation or survival. Combining integration site analysis and bulk RNA sequencing, we established that an upregulated STAT3 signature due to proviral insertional mutagenesis was associated with persistent HIV-infected CD4+ T cells. HIV+ persistent cells also expressed a STAT3-related anti-apoptotic and cytotoxic phenotype that resembles that of HIV-associated T cell lymphomas. HIV insertional mutagenesis of STAT3 and expression of its downstream targets provides a model of HIV-associated T cell lymphomas that can be used to further determine the oncogenic drivers of HIV-associated lymphomas, both AIDS- and gene therapy-associated, and, potentially, to evaluate therapeutics against these HIV-associated cancers. Author Summary The effects of HIV proviral insertional mutagenesis have been demonstrated in a handful of HIV-associated T cell lymphomas, where integration of an HIV provirus within intron 1 of STAT3 , results in increased expression of the STAT3 protein. To study the effects of HIV insertional mutagenesis, we established an ex vivo culture protocol of primary human CD4+ T cells infected with a replication-incompetent HIV vector with a gfp-reporter. After infection, the HIV/GFP+ cells from all three donors declined, but, over time, 3/6 replicates from one donor populations of infected cells rebounded. The resurgent HIV/GFP+ cells contained a provirus integrated within intron 1 of STAT3 , which led to increases in gene expression, STAT3 activation, and upregulation of a STAT3 -associated anti-apoptotic and cytotoxic phenotype. The STAT3 -associated gene signature shared similarities to the HIV-associated lymphomas with similar integration sites. Additionally, in all 3 replicates, insertional mutagenesis of genes other than STAT3 may have also contributed to clonal expansion of HIV/GFP+ T cells. Overall, we have demonstrated that HIV provirus insertional mutagenesis can influence T cell persistence. Our study provides a primary T cell culture model system that can be used to further study how proviral insertional mutagenesis influences HIV-associated T cell lymphomas and the safety of lentiviral vectors used in gene and cell therapies.
Collapse
|
13
|
Chen Z, Qin Y. Role of miRNA‑145‑5p in cancer (Review). Oncol Rep 2025; 53:39. [PMID: 39886965 PMCID: PMC11800069 DOI: 10.3892/or.2025.8872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
MicroRNA‑145‑5p (miRNA‑145‑5p) is a short non‑coding RNA located at chromosome 5q33.1, which has gained significant attention in several aspects of cellular regulation and biological functions. In malignant tumours, miRNA‑145‑5p may function as either a tumour suppressor or an oncogene, affecting tumour progression by targeting downstream genes or modulating their expression through upstream regulators. However, the full extent of miRNA‑145‑5p's role in cancer has remained to be determined. This review provides an overview of the role of miRNA‑145‑5p in cancer, investigates its potential as a biomarker for diagnosis, prognosis and treatment response, and evaluates its influence on cancer chemotherapy and radiotherapy. Finally, current strategies for systemic delivery of miRNA‑145‑5p in cancer therapies are summarized.
Collapse
Affiliation(s)
- Zeshan Chen
- Department of Traditional Chinese Medicine, Guangxi Zhuang Autonomous Region People's Hospital, Nanning, Guangxi 530016, P.R. China
| | - Yijue Qin
- Department of Traditional Chinese Medicine, Guangxi Zhuang Autonomous Region People's Hospital, Nanning, Guangxi 530016, P.R. China
| |
Collapse
|
14
|
Gonzalez JC, Park KW, Evans DB, Sharma R, Sahaym O, Gopalakrishnan S, dar AI, Valdez TA, Sharma A. Nano Approaches to Nucleic Acid Delivery: Barriers, Solutions, and Current Landscape. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70010. [PMID: 40223402 PMCID: PMC11994986 DOI: 10.1002/wnan.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 03/07/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
Nucleic acid (NA) therapy holds tremendous potential for treating a wide range of genetic diseases by the delivery of therapeutic genes into target cells. However, significant challenges exist in safely and effectively delivering these genes to their intended locations. Viral vectors, though efficient, pose risks such as immunogenicity and mutagenesis. This has resulted in growing interest in non-viral, nanoparticle-based NA delivery systems. This review article describes various physiological barriers to NA delivery and explores nanoparticle-based NA delivery systems, including bioengineered nanoparticles, peptides, lipid nanoparticles, and polymeric nanoparticles, highlighting their unique features to overcome in vivo barriers for NA delivery. While these nanoparticle-based NA delivery systems offer a promising alternative to viral vectors, challenges related to cytotoxicity, reproducible synthesis, and cost need to be addressed. The current clinical landscape of NA delivery is also discussed, emphasizing the need for safer, scalable, and cost-effective solutions. Nanoparticles represent a promising future in NA therapy, with the possibility of developing clinically relevant, non-toxic, stable, and non-immunogenic delivery vehicles, paving the way for broader therapeutic applications and improved clinical outcomes.
Collapse
Affiliation(s)
- Joan Castaneda Gonzalez
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Ki Wan Park
- Department of Otolaryngology−Head & Neck Surgery DivisionsStanford University School of MedicineStanfordCaliforniaUSA
| | - Dallin Brian Evans
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Rishi Sharma
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Om Sahaym
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Shamila Gopalakrishnan
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Aqib Iqbal dar
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| | - Tulio A. Valdez
- Department of Otolaryngology−Head & Neck Surgery DivisionsStanford University School of MedicineStanfordCaliforniaUSA
| | - Anjali Sharma
- Department of ChemistryCollege of Arts and Sciences, Washington State UniversityPullmanWashingtonUSA
| |
Collapse
|
15
|
Dong J, Wu J, Jin Y, Zheng Z, Su T, Shao L, Bei J, Chen S. In-depth analysis of the safety of CAR-T cell therapy for solid tumors. Front Immunol 2025; 16:1548979. [PMID: 40066440 PMCID: PMC11891211 DOI: 10.3389/fimmu.2025.1548979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/30/2025] [Indexed: 05/13/2025] Open
Abstract
In recent years, the rapid progress in oncology, immunology, and molecular biology has dramatically advanced cancer immunotherapy, particularly CAR-T cell therapy. This innovative approach involves engineering a patient's T cells to express receptors that specifically target tumor antigens, enhancing their ability to identify and eliminate cancer cells. However, the effectiveness of CAR-T therapy in solid tumors is often hampered by the challenging tumor microenvironment (TME). The complex TME includes dense stroma that obstructs T cell infiltration, abnormal blood vessel structures leading to hypoxia, and an acidic pH, all of which hinder CAR-T cell function. Additionally, the presence of immunosuppressive factors in the TME reduces the efficacy of CAR-T cells, making successful targeting of tumors more difficult. The safety of CAR-T therapy has gained interest, especially CAR-T therapy has shown considerable effectiveness in various cancers, with notable results in multiple myeloma and hepatocellular carcinoma, among others. Nonetheless, CAR-T cell therapy is associated with several adverse reactions primarily driven by heightened levels of proinflammatory cytokines. These reactions include cytokine release syndrome (CRS), neurotoxicity (CANS), and organ toxicity, often leading to serious complications. CRS, characterized by systemic inflammation due to cytokine release, can escalate to severe organ dysfunction. It typically occurs within the first week post-infusion, correlating with CAR-T cell expansion and often presents with fever and hypotension. Meanwhile, CANS encompasses neurological issues ranging from mild symptoms to severe seizures, possibly exacerbated by CRS. Organ toxicity can also arise from CAR-T therapy, with potential damage affecting the gastrointestinal tract, kidneys, liver, and lungs, often tied to shared antigens found in both tumor and healthy tissues. Moreover, long-term effects like cytokine-associated hematotoxicity (CAHT) and secondary malignancies represent significant concerns that could affect the patient's quality of life post-treatment. The long-term adverse effects and challenges in treating solid tumors underscore the need for ongoing research. Strategies to improve CAR-T cell efficacy, minimize adverse reactions, and enhance patient safety are critical. Future explorations could include designing CAR-T cells to better navigate the TME, identifying specific target antigen profiles to minimize off-target damage, and developing adjunct therapies to mitigate cytokine-related toxicity. Continued monitoring for long-term effects will also be paramount in improving patient outcomes and maintaining their quality of life. Overall, while CAR-T therapy holds great promise, it must be administered with careful consideration of potential side effects and rigorous management strategies to ensure patient safety and treatment efficacy.
Collapse
Affiliation(s)
- Jiayi Dong
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cancer Immunotherapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiexiong Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ye Jin
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cancer Immunotherapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhu Zheng
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cancer Immunotherapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ting Su
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cancer Immunotherapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lijuan Shao
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cancer Immunotherapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiaxin Bei
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cancer Immunotherapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
| | - Size Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Cancer Immunotherapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
16
|
Kamenšek U, Božič T, Čemažar M, Švajger U. Antitumor Efficacy of Interleukin 12-Transfected Mesenchymal Stem Cells in B16-F10 Mouse Melanoma Tumor Model. Pharmaceutics 2025; 17:278. [PMID: 40142942 PMCID: PMC11944637 DOI: 10.3390/pharmaceutics17030278] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Mesenchymal stromal cells (MSCs) hold the potential for tumor-targeted gene delivery due to their ex vivo manipulability, low immunogenicity, scalability, and inherent tumor-homing properties. Despite the widespread use of viral vectors for MSC genetic modification, safety concerns have prompted interest in non-viral alternatives, such as gene electrotransfer (GET). This study aimed to optimize GET parameters for MSCs transfection, assess MSCs biodistribution after in vivo administration, and evaluate the therapeutic potential of interleukin-12 (IL-12)-modified MSCs in a mouse melanoma model. Methods: Human MSCs were isolated from umbilical cords under ethically approved protocols. GET protocols were optimized using a fluorescent reporter gene to evaluate transfection efficiency and cell viability. MSC biodistribution was examined following intravenous and intratumoral injections in murine tumor models using luminescent reporter gene. The therapeutic efficacy of IL-12-modified MSCs was assessed in a syngeneic mouse melanoma model. Results: Optimized GET protocols achieved a transfection efficiency of 80% and a cell viability of 90%. Biodistribution studies demonstrated effective tumor retention of MSCs following intratumoral injections, whereas intravenous administration resulted in predominant cell localization in the lungs. IL-12-modified MSCs injected intratumorally significantly inhibited tumor growth, delaying tumor progression by five days compared to controls. Conclusions: Optimized GET conditions enabled high-efficiency, high-viability MSCs transfection, facilitating their use as effective vehicles for localized cytokine delivery. While the innate tumor tropism of MSCs was not conclusively demonstrated, the study highlights the potential of GET as a reliable non-viral gene delivery platform and underscores the therapeutic promise of IL-12-modified MSCs in tumor-targeted gene therapy.
Collapse
Affiliation(s)
- Urška Kamenšek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, SI-1000 Ljubljana, Slovenia; (U.K.); (T.B.)
- Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Tim Božič
- Department of Experimental Oncology, Institute of Oncology Ljubljana, SI-1000 Ljubljana, Slovenia; (U.K.); (T.B.)
| | - Maja Čemažar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, SI-1000 Ljubljana, Slovenia; (U.K.); (T.B.)
- Biotechnical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, SI-6310 Izola, Slovenia
| | - Urban Švajger
- Department for Therapeutic Services, Slovenian Institute for Transfusion Medicine, SI-1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
17
|
Ressnerova A, Heger Z, Pumera M. Translational nanorobotics breaking through biological membranes. Chem Soc Rev 2025; 54:1924-1956. [PMID: 39807638 DOI: 10.1039/d4cs00483c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
In the dynamic realm of translational nanorobotics, the endeavor to develop nanorobots carrying therapeutics in rational in vivo applications necessitates a profound understanding of the biological landscape of the human body and its complexity. Within this landscape, biological membranes stand as critical barriers to the successful delivery of therapeutic cargo to the target site. Their crossing is not only a challenge for nanorobotics but also a pivotal criterion for the clinical success of therapeutic-carrying nanorobots. Nevertheless, despite their urgency, strategies for membrane crossing in translational nanorobotics remain relatively underrepresented in the scientific literature, signaling an opportunity for further research and innovation. This review focuses on nanorobots with various propulsion mechanisms from chemical and physical to hybrid mechanisms, and it identifies and describes four essential biological membranes that represent the barriers needed to be crossed in the therapeutic journey of nanorobots in in vivo applications. First is the entry point into the blood stream, which is the skin or mucosa or intravenous injection; next is the exit from the bloodstream across the endothelium to the target site; further is the entry to the cell through the plasma membrane and, finally, the escape from the lysosome, which otherwise destroys the cargo. The review also discusses design challenges inherent in translating nanorobot technologies to real-world applications and provides a critical overview of documented membrane crossings. The aim is to underscore the need for further interdisciplinary collaborations between chemists, materials scientists and chemical biologists in this vital domain of translational nanorobotics that has the potential to revolutionize the field of precision medicine.
Collapse
Affiliation(s)
- Alzbeta Ressnerova
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00, Brno, Czech Republic.
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
| | - Zbynek Heger
- Research Group for Molecular Biology and Nanomedicine, Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
- Center of Advanced Innovation Technologies, Faculty of Materials Science and Technology, VSB - Technical University of Ostrava, 17. Listopadu 2172/15, 70800 Ostrava, Czech Republic
| | - Martin Pumera
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-612 00, Brno, Czech Republic.
- Advanced Nanorobots & Multiscale Robotics Laboratory, Faculty of Electrical Engineering and Computer Science, VSB - Technical University of Ostrava, 17. listopadu 2172/15, 70800 Ostrava, Czech Republic
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
- Department of Medical Research, China Medical University Hospital, China Medical University, No. 91 Hsueh-Shih Road, Taichung, Taiwan
| |
Collapse
|
18
|
Holzwarth D, Calaminus G, Friese J, Sejersen T, Büning H, John-Neek P, Bastone AL, Rothe M, Mansfield K, Libertini S, Dubost V, Kuzmiski B, Alecu I, Labik I, Kirschner J. Pilocytic astrocytoma in a child with spinal muscular atrophy treated with onasemnogene abeparvovec. Mol Ther 2025:S1525-0016(25)00115-7. [PMID: 39955617 DOI: 10.1016/j.ymthe.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/21/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a severe neuromuscular disease, leading to progressive muscle weakness and potentially early mortality if untreated. Onasemnogene abeparvovec is a recombinant adeno-associated virus serotype 9 (rAAV9)-based gene therapy that has demonstrated improvements in survival and motor function for SMA patients. Here, we present a case of a patient diagnosed with a grade 1 pilocytic astrocytoma at the age of 2 years, approximately 8 months after onasemnogene abeparvovec treatment. Although vector genomes delivered by rAAVs persist primarily as episomes, rare integration events have been linked to tumor formation in neonate murine models. Therefore, we investigated the presence and possible integration of onasemnogene abeparvovec in formalin-fixed paraffin embedded (FFPE) and frozen tumor samples. In situ hybridization demonstrated variable transduction levels in individual tumor cells, while droplet digital PCR measured an average vector copy number ranging from 0.7 to 4.9 vector genomes/diploid genome. Integration site analysis identified a low number of integration sites that were not conserved between technical replicates, nor between FFPE and frozen samples, indicating that cells hosting integrating vector genomes represented a minority in the overall cell population. Thus, molecular analysis of the tumor tissue suggests that tumorigenesis was causally independent of the administration of onasemnogene abeparvovec.
Collapse
Affiliation(s)
| | | | | | - Thomas Sejersen
- Pediatric Neurology, Karolinska University Hospital, 171 77 Stockholm, Sweden; Department of Women's and Children's Health, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Philipp John-Neek
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | | | | | | | | | - Iulian Alecu
- Novartis Pharmaceuticals, 4056 Basel, Switzerland
| | - Ivan Labik
- ProtaGene CGT GmbH, 69120 Heidelberg, Germany
| | - Janbernd Kirschner
- Department for Neuropediatrics and Muscle Disease, Medical Center - University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
| |
Collapse
|
19
|
Guthrie OW. Gene Therapy: An Historical Overview for Familial Hearing Loss. Int J Mol Sci 2025; 26:1469. [PMID: 40003934 PMCID: PMC11855000 DOI: 10.3390/ijms26041469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Gene therapy is a promising molecular approach for the management of familial hearing loss. This type of molecular therapy is the physical manifestation of genetic determinism-the notion that individual genes cause individual phenotypes. The current composition weaves through various branches of the biomedical sciences to uncover the molecular biologic premise for genetic determinism and the impetus behind gene therapy. Consequently, it is revealed that the underlying molecular biologic premise was scaffolded on successful observations from simple biologic assays that were devoid of the complexities of human disease biology. Furthermore, modern successful gene therapies are largely driven by commercial and academic incentives at the cost of scientific rigor. This poses several perverse challenges for patients, clinicians and the public at large. Issues concerning safety, efficacy, and ethics are far from resolved despite regulatory agency approvals, the media's bias for gene therapy and the many lucrative investor positions. Lastly, the therapeutic claims regarding gene therapy are the most ambitious claims made within the hearing sciences. Therefore, scientists, clinicians, and patients must be equipped with the tools needed to appropriately consume and appraise such claims. These and other issues are also directly addressed, with the aim of providing a realistic sense of whether current human gene therapies are ready to be positioned within our routine clinical armamentarium against hearing loss.
Collapse
Affiliation(s)
- O'neil W Guthrie
- Cell & Molecular Pathology Laboratory, Department of Communication Sciences and Disorders, Northern Arizona University, Flagstaff, AZ 86011, USA
| |
Collapse
|
20
|
Berg P, Ruppert-Seipp G, Müller S, Maurer GD, Hartmann J, Holtick U, Buchholz CJ, Funk MB. CAR T-cell-associated secondary malignancies challenge current pharmacovigilance concepts. EMBO Mol Med 2025; 17:211-218. [PMID: 39762651 PMCID: PMC11822130 DOI: 10.1038/s44321-024-00183-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 02/14/2025] Open
Abstract
Suspected adverse reactions following chimeric antigen receptor T-cell (CAR T) treatment include more and more cases of secondary T-cell malignancies. The causality assessment of such suspected reactions challenges established evaluation practices due to (i) patient and product-specific risk factors and (ii) incomplete data available with post-marketing reports submitted to competent authorities. This is of particular relevance for gene therapy products that integrate into the host genome. We present a summary of case reports related to different CAR T products and the rationale for case causality assessment. In this context, possible pathophysiologic mechanisms and differences between CAR T products to be taken into account are discussed. The unparalleled complexity of the case follow-up and the multistep process of cancer development necessitates a case-by-case consideration. This highlights challenges in the pharmacovigilance of advanced therapy medicinal products and underlines the importance of testing for vector presence, integration location and gene expression profile for an informed case assessment of suspected secondary malignancies with the aim to obtain a better understanding of contributing factors.
Collapse
Affiliation(s)
- Philipp Berg
- Safety of Biomedicines and Diagnostics, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Susanne Müller
- Safety of Biomedicines and Diagnostics, Paul-Ehrlich-Institut, Langen, Germany
| | - Gabriele D Maurer
- Safety of Biomedicines and Diagnostics, Paul-Ehrlich-Institut, Langen, Germany
| | - Jessica Hartmann
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany.
| | - Markus B Funk
- Safety of Biomedicines and Diagnostics, Paul-Ehrlich-Institut, Langen, Germany.
| |
Collapse
|
21
|
Oliveira BC, Bari S, Melenhorst JJ. Leveraging Vector-Based Gene Disruptions to Enhance CAR T-Cell Effectiveness. Cancers (Basel) 2025; 17:383. [PMID: 39941752 PMCID: PMC11815729 DOI: 10.3390/cancers17030383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy represents a breakthrough in the treatment of relapsed and refractory B-cell malignancies, such as chronic lymphocytic leukemia (CLL), inducing long-term, sometimes curative, responses. However, fewer than 30% of CLL patients achieve such outcomes. It has been shown that a smaller subset of T cells capable of expansion and persistence is crucial for treatment effectiveness. Notably, a pre-existing mutation in the epigenetic regulator TET2, combined with CAR vector-induced disruption of the other intact allele, significantly enhanced the potency of the CAR-engineered T-cell clone in one CLL patient. This finding aligns with independent research, suggesting that the CAR gene's genomic insertion site influences tumor-targeting capability. Thus, it is plausible that vector-induced gene disruptions affect CAR T-cell function. This review synthesizes existing knowledge on vector integration into the host genome and its impact on clinical outcomes in CAR T-cell therapy patients. Our aim is to inform the development of improved therapies and enhance their overall efficacy.
Collapse
Affiliation(s)
| | | | - J. Joseph Melenhorst
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44016, USA; (B.C.O.); (S.B.)
| |
Collapse
|
22
|
Inamdar A, Gurupadayya B, Halagali P, Nandakumar S, Pathak R, Singh H, Sharma H. Cutting-edge Strategies for Overcoming Therapeutic Barriers in Alzheimer's Disease. Curr Pharm Des 2025; 31:598-618. [PMID: 39492772 DOI: 10.2174/0113816128344571241018154506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) remains one of the hardest neurodegenerative diseases to treat due to its enduring cognitive deterioration and memory loss. Despite extensive research, few viable treatment approaches have been found; these are mostly due to several barriers, such as the disease's complex biology, limited pharmaceutical efficacy, and the BBB. This presentation discusses current strategies for addressing these therapeutic barriers to enhance AD treatment. Innovative drug delivery methods including liposomes, exosomes, and nanoparticles may be able to pass the blood-brain barrier and allow medicine to enter specific brain regions. These innovative strategies of medicine distribution reduce systemic side effects by improving absorption. Moreover, the development of disease-modifying treatments that target tau protein tangles, amyloid-beta plaques, and neuroinflammation offers the chance to influence the course of the illness rather than only treat its symptoms. Furthermore, gene therapy and CRISPR-Cas9 technologies have surfaced as potentially groundbreaking methods for addressing the underlying genetic defects associated with AD. Furthermore, novel approaches to patient care may involve the utilization of existing medications having neuroprotective properties, such as those for diabetes and cardiovascular conditions. Furthermore, biomarker research and personalized medicine have made individualized therapy approaches possible, ensuring that patients receive the best care possible based on their unique genetic and molecular profiles.
Collapse
Affiliation(s)
- Aparna Inamdar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Bannimath Gurupadayya
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Prashant Halagali
- Department of Pharmaceutical Quality Assurance, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi 590010, Karnataka, India
| | - S Nandakumar
- Associate Scientist, Corteva Agriscience, Hyderabad 500081, Telangana, India
| | - Rashmi Pathak
- Department of Pharmacy, Invertis University, Bareilly (UP) 243123, India
| | - Himalaya Singh
- Department of Medicine, Government Institute of Medical Sciences, Greater Noida (UP) 201312, India
| | - Himanshu Sharma
- Department of Pharmacy, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad (UP) 244001, India
| |
Collapse
|
23
|
Moiseenko A, Sinadinos A, Sergijenko A, Pineault K, Saleh A, Nekola K, Strang N, Eleftheraki A, Boyd AC, Davies JC, Gill DR, Hyde SC, McLachlan G, Rath T, Rothe M, Schambach A, Hobbie S, Schuler M, Maier U, Thomas MJ, Mennerich D, Schmidt M, Griesenbach U, Alton EWFW, Kreuz S. Pharmacological and pre-clinical safety profile of rSIV.F/HN, a hybrid lentiviral vector for cystic fibrosis gene therapy. Eur Respir J 2025; 65:2301683. [PMID: 39174284 PMCID: PMC11780724 DOI: 10.1183/13993003.01683-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 08/02/2024] [Indexed: 08/24/2024]
Abstract
RATIONALE AND OBJECTIVE Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. CFTR modulators offer significant improvements, but ∼10% of patients remain nonresponsive or are intolerant. This study provides an analysis of rSIV.F/HN, a lentiviral vector optimised for lung delivery, including CFTR protein expression, functional correction of CFTR defects and genomic integration site analysis in preparation for a first-in-human clinical trial. METHODS Air-liquid interface cultures of primary human bronchial epithelial cells (HBECs) from CF patients (F508del/F508del), as well as a CFTR-deficient immortalised human lung epithelial cell line mimicking class I (CFTR-null) homozygous mutations, were used to assess transduction efficiency. Quantification methods included a novel proximity ligation assay for CFTR protein expression. For assessment of CFTR channel activity, Ussing chamber studies were conducted. The safety profile was assessed using integration site analysis and in vitro insertional mutagenesis studies. RESULTS rSIV.F/HN expressed CFTR and restored CFTR-mediated chloride currents to physiological levels in primary F508del/F508del HBECs as well as in a class I cells. In contrast, the latter could not be achieved by small-molecule CFTR modulators, underscoring the potential of gene therapy for this mutation class. Combination of rSIV.F/HN-CFTR with the potentiator ivacaftor showed a greater than additive effect. The genomic integration pattern showed no site predominance (frequency of occurrence ≤10%), and a low risk of insertional mutagenesis was observed in an in vitro immortalisation assay. CONCLUSIONS The results underscore rSIV.F/HN as a promising gene therapy vector for CF, providing a mutation-agnostic treatment option.
Collapse
Affiliation(s)
- Alena Moiseenko
- Boehringer Ingelheim Pharma GmbH, Biberach an der Riss, Germany
| | - Anthony Sinadinos
- UK Respiratory Gene Therapy Consortium, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ana Sergijenko
- UK Respiratory Gene Therapy Consortium, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kyriel Pineault
- UK Respiratory Gene Therapy Consortium, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Aarash Saleh
- UK Respiratory Gene Therapy Consortium, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Konradin Nekola
- Boehringer Ingelheim Pharma GmbH, Biberach an der Riss, Germany
| | - Nathalie Strang
- Boehringer Ingelheim Pharma GmbH, Biberach an der Riss, Germany
| | | | - A Christopher Boyd
- UK Respiratory Gene Therapy Consortium, London, UK
- Centre of Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jane C Davies
- UK Respiratory Gene Therapy Consortium, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Depts of Respiratory Medicine and Paediatric Respiratory Medicine, Royal Brompton Hospital, Guy's and St Thomas' Trust, London, UK
| | - Deborah R Gill
- UK Respiratory Gene Therapy Consortium, London, UK
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen C Hyde
- UK Respiratory Gene Therapy Consortium, London, UK
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Gerry McLachlan
- UK Respiratory Gene Therapy Consortium, London, UK
- The Roslin Institute & R(D)SVS, University of Edinburgh, Edinburgh, UK
| | - Tim Rath
- ProtaGene CGT (former GeneWerk GmbH), Heidelberg, Germany
| | | | - Axel Schambach
- Medizinische Hochschule Hannover, Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Silke Hobbie
- Boehringer Ingelheim Pharma GmbH, Biberach an der Riss, Germany
| | - Michael Schuler
- Boehringer Ingelheim Pharma GmbH, Biberach an der Riss, Germany
| | - Udo Maier
- Boehringer Ingelheim Pharma GmbH, Biberach an der Riss, Germany
| | | | | | - Manfred Schmidt
- ProtaGene CGT (former GeneWerk GmbH), Heidelberg, Germany
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Deceased
| | - Uta Griesenbach
- UK Respiratory Gene Therapy Consortium, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- U. Griesenbach, E.W.F.W. Alton and S. Kreuz are joint senior authors
| | - Eric W F W Alton
- UK Respiratory Gene Therapy Consortium, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Depts of Respiratory Medicine and Paediatric Respiratory Medicine, Royal Brompton Hospital, Guy's and St Thomas' Trust, London, UK
- U. Griesenbach, E.W.F.W. Alton and S. Kreuz are joint senior authors
| | - Sebastian Kreuz
- Boehringer Ingelheim Pharma GmbH, Biberach an der Riss, Germany
- U. Griesenbach, E.W.F.W. Alton and S. Kreuz are joint senior authors
| |
Collapse
|
24
|
Corydon TJ, Bek T. Multiple gene therapy as a tool for regulating the expression of molecules involved in neovascular age-related macular degeneration. Prog Retin Eye Res 2025; 104:101323. [PMID: 39672501 DOI: 10.1016/j.preteyeres.2024.101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/08/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Anti-vascular endothelial growth factor (VEGF) therapies have revolutionized the treatment of neovascular age-related macular degeneration (nAMD) and other retinal diseases. However, the necessity for repeated intravitreal injections and the observation of variable treatment responses calls for new treatment modalities where fewer and more effective interventions can result in a clinical effect. Gene therapy might be such an alternative, and therefore the development and clinical application of gene therapy aimed at modifying gene expression has received considerable attention. The article reviews current knowledge of the background, pathophysiological mechanisms, technologies, limitations, and future directions for gene therapy aimed at modifying the synthesis of compounds involved in acquired and senescent retinal disease. The authors have contributed to the field by developing gene therapy to reduce the expression of vascular endothelial growth factor (VEGF), as well as multiple gene therapy for simultaneous downregulation of the synthesis of VEGF and upregulation of pigment epithelium-derived factor (PEDF) using adeno-associated virus (AAV) vectors. It is suggested that such multi-target gene therapy might be included in future treatments of retinal diseases where the underlying mechanisms are complex and cannot be attributed to one specific mediator. Such diseases might include dry AMD (dAMD) with geographic atrophy, but also diabetic macular edema (DME) and retinal vein occlusion (RVO). Gene therapy can be expected to be most beneficial for the patients in need of multiple intra-vitreal injections and in whom the therapeutic response is insufficient. It is concluded, that in parallel with basic research, there is a need for clinical studies aimed at identifying factors that can be used to identify patients who will benefit from gene therapy already at the time of diagnosis of the retinal disease.
Collapse
Affiliation(s)
- Thomas J Corydon
- Department of Biomedicine, Hoegh Guldbergs Gade 10, Aarhus University, 8000, Aarhus C, Denmark; Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
25
|
Brusson M, Miccio A. [A CRISPR/Cas approach to β-haemoglobinopathies]. Med Sci (Paris) 2025; 41:33-39. [PMID: 39887096 DOI: 10.1051/medsci/2024191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025] Open
Abstract
Beta-haemoglobinopathies are severe genetic anemias caused by mutations that affect adult haemoglobin production. Many therapeutic approaches aim to reactivate the expression of the fetal hemoglobin genes. To this end, the CRISPR/Cas9 system has recently been used to genetically modify patients' hematopoietic stem/progenitor cells ex vivo and reactivate fetal hemoglobin expression in their erythroid progeny. More than 70 patients with severe β-thalassemia and sickle cell disease have been treated with the Casgevy® therapy. Most have achieved a significant improvement of clinical phenotype, with high editing efficiency in hematopoietic cells associated with normal or near normal hemoglobin levels. While the long-term safety and efficacy of this powerful approach still need to be evaluated, new strategies are being developed to further improve therapeutic outcomes, reduce potential genotoxicity and lower the costs of therapy.
Collapse
Affiliation(s)
- Megane Brusson
- Institut Imagine, Inserm UMR1163, université Paris Cité, Paris, France
| | - Annarita Miccio
- Institut Imagine, Inserm UMR1163, université Paris Cité, Paris, France
| |
Collapse
|
26
|
Murphy R, Martin KR. Genetic engineering and the eye. Eye (Lond) 2025; 39:57-68. [PMID: 39516652 PMCID: PMC11733221 DOI: 10.1038/s41433-024-03441-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 07/22/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The transformative potential of genetic engineering in ophthalmology is remarkable, promising new treatments for a wide range of blinding eye diseases. The eye is an attractive target organ for genetic engineering approaches, in part due to its relatively immune-privileged status, its accessibility, and the ease of monitoring of efficacy and safety. Consequently, the eye has been at the forefront of genetic engineering advances in recent years. The development of Clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9), base editors, prime editors, and transposases have enabled efficient and specific gene modification. Ocular gene therapy continues to progress, with recent advances in delivery systems using viral / non-viral vectors and novel promoters and enhancers. New strategies to achieve neuroprotection and neuroregeneration are evolving, including direct in-vivo cell reprogramming and optogenetic approaches. In this review, we discuss recent advances in ocular genetic engineering, examine their current therapeutic roles, and explore their potential use in future strategies to reduce the growing burden of vision loss and blindness.
Collapse
Affiliation(s)
- Rory Murphy
- Department of Ophthalmology, Royal Victoria Eye and Ear Hospital, Dublin, Ireland
- Department of Ophthalmology, Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia
| | - Keith R Martin
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia.
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
27
|
Ballantine J, Tisdale JF. Gene therapy for sickle cell disease: recent advances, clinical trials and future directions. Cytotherapy 2024:S1465-3249(24)00925-3. [PMID: 39729054 DOI: 10.1016/j.jcyt.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/03/2024] [Indexed: 12/28/2024]
Abstract
Sickle cell disease (SCD) is the most common inherited blood disorder worldwide, impacting millions and imposing severe healthcare challenges, particularly in resource-limited regions. Current treatments have variable efficacy and require lifelong adherence. Allogeneic Hematopoietic Stem Cell Transplantation can be curative but comes with significant side effects and limited donor availability limits its widespread applicability. Gene therapy, by addressing the root genetic causes, offers a revolutionary alternative. This article discusses the molecular mechanisms of SCD and β-thalassemia and highlights advancements in gene therapy, such as gene addition via lentiviral vectors and gene editing with CRISPR/Cas9 technology. Clinical trials have brought about significant progress but challenges remain, including leukemogenesis, delivery efficiency and cost. Future efforts must focus on enhancing efficiency, reducing costs, developing nongenotoxic conditioning regimens and methods for in vivo application.
Collapse
Affiliation(s)
- Josiah Ballantine
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA.
| | - John F Tisdale
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
28
|
Kim Y, Landstrom AP, Shah SH, Wu JC, Seidman CE. Gene Therapy in Cardiovascular Disease: Recent Advances and Future Directions in Science: A Science Advisory From the American Heart Association. Circulation 2024; 150:e471-e480. [PMID: 39523949 DOI: 10.1161/cir.0000000000001296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Cardiovascular disease remains the foremost cause of morbidity and mortality globally, affecting millions of individuals. Recent discoveries illuminate the substantial role of genetics in cardiovascular disease pathogenesis, encompassing both monogenic and polygenic mechanisms and identifying tangible targets for gene therapies. Innovative strategies have emerged to rectify pathogenic variants that cause monogenic disorders such as hypertrophic, dilated, and arrhythmogenic cardiomyopathies and hypercholesterolemia. These include delivery of exogenous genes to supplement insufficient protein levels caused by pathogenic variants or genome editing to correct, delete, or modify mutant sequences to restore protein function. However, effective delivery of gene therapy to specified cells presents formidable challenges. Viral vectors, notably adeno-associated viruses and nonviral vectors such as lipid and engineered nanoparticles, offer distinct advantages and limitations. Additional risks and obstacles remain, including treatment durability, tissue-specific targeting, vector-associated adverse events, and off-target effects. Addressing these challenges is an ongoing imperative; several clinical gene therapy trials are underway, and many more first-in-human studies are anticipated. This science advisory reviews core concepts of gene therapy, key obstacles, patient risks, and ongoing research endeavors to enable clinicians to understand the complex landscape of this emerging therapy and its remarkable therapeutic potential to benefit cardiovascular disease.
Collapse
|
29
|
Zhang H, Wei Y, Wang Y, Liang J, Hou Y, Nie X, Hou J. Emerging Diabetes Therapies: Regenerating Pancreatic β Cells. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:644-656. [PMID: 39276101 DOI: 10.1089/ten.teb.2024.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
The incidence of diabetes mellitus (DM) is steadily increasing annually, with 537 million diabetic patients as of 2021. Restoring diminished β cell mass or impaired islet function is crucial in treating DM, particularly type 1 DM. However, the regenerative capacity of islet β cells, which primarily produce insulin, is severely limited, and natural regeneration is only observed in young rodents or children. Hence, there is an urgent need to develop advanced therapeutic approaches that can regenerate endogenous β cells or replace them with stem cell (SC)-derived or engineered β-like cells. Current strategies for treating insulin-dependent DM mainly include promoting the self-replication of endogenous β cells, inducing SC differentiation, reprogramming non-β cells into β-like cells, and generating pancreatic-like organoids through cell-based intervention. In this Review, we discuss the current state of the art in these approaches, describe associated challenges, propose potential solutions, and highlight ongoing efforts to optimize β cell or islet transplantation and related clinical trials. These effective cell-based therapies will generate a sustainable source of functional β cells for transplantation and lay strong foundations for future curative treatments for DM.
Collapse
Affiliation(s)
- Haojie Zhang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yaxin Wei
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yubo Wang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jialin Liang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yifan Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng, China
- Department of Urinary Surgery, Henan Provincial Research Center for the Prevention and Diagnosis of Prostate Diseases, Huaihe Hospital, Henan University, Kaifeng, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Department of Urinary Surgery, Henan Provincial Research Center for the Prevention and Diagnosis of Prostate Diseases, Huaihe Hospital, Henan University, Kaifeng, China
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng, China
| |
Collapse
|
30
|
Hosuru RV, Yang J, Zhou Y, Gin A, Hayal TB, Hong SG, Dunbar CE, Wu C. Long-term tracking of haematopoietic clonal dynamics and mutations in non-human primate undergoing transplantation of lentivirally barcoded haematopoietic stem and progenitor cells. Br J Haematol 2024; 205:2487-2497. [PMID: 39523608 PMCID: PMC11637732 DOI: 10.1111/bjh.19889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Haematopoietic stem and progenitor cell (HSPC) autologous gene therapies are promising treatment for a variety of blood disorders. Investigation of the long-term HSPC clonal dynamics and other measures of safety and durability following lentiviral-mediated gene therapies in predictive models are crucial for assessing risks and benefits in order to inform decisions regarding wider utilization. We established an autologous lentivirally barcoded HSPC transplantation model in rhesus macaque (RM), a model offering insights into haematopoiesis and gene therapies with direct relevance to human. Healthy young adult RMs underwent total body irradiation, followed by transplantation of autologous HSPCs transduced with a lentiviral vector containing a diverse genetic barcode library, uniquely labelling individual HSPCs and their progeny. With up to 131 months of follow-up, we now report quantitative clonal dynamics, characterizing the number, diversity, stability and lineage bias of hundreds of thousands of HSPC clones tracked in five RMs. We documented long-term stable and multi-lineage output from a highly polyclonal pool of HSPCs. Clonal succession after stable haematopoietic reconstitution was minimal. There was no evidence for accelerated acquisition of acquired somatic mutations following autologous lentivirally transduced HSPC transplantation. Our results provide relevant insights into long-term HSPC behaviours in vivo following transplantation and gene therapies.
Collapse
Affiliation(s)
- Rohan V. Hosuru
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Jack Yang
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Yifan Zhou
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeUK
- Haematological Cancer GeneticsWellcome Trust Sanger InstituteCambridgeUK
| | - Ashley Gin
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Taha B. Hayal
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - So Gun Hong
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Cynthia E. Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Chuanfeng Wu
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
31
|
Gilioli G, Lankester AC, de Kivit S, Staal FJT, Ott de Bruin LM. Gene therapy strategies for RAG1 deficiency: Challenges and breakthroughs. Immunol Lett 2024; 270:106931. [PMID: 39303994 DOI: 10.1016/j.imlet.2024.106931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Mutations in the recombination activating genes (RAG) cause various forms of immune deficiency. Hematopoietic stem cell transplantation (HSCT) is the only cure for patients with severe manifestations of RAG deficiency; however, outcomes are suboptimal with mismatched donors. Gene therapy aims to correct autologous hematopoietic stem and progenitor cells (HSPC) and is emerging as an alternative to allogeneic HSCT. Gene therapy based on viral gene addition exploits viral vectors to add a correct copy of a mutated gene into the genome of HSPCs. Only recently, after a prolonged phase of development, viral gene addition has been approved for clinical testing in RAG1-SCID patients. In the meantime, a new technology, CRISPR/Cas9, has made its debut to compete with viral gene addition. Gene editing based on CRISPR/Cas9 allows to perform targeted genomic integrations of a correct copy of a mutated gene, circumventing the risk of virus-mediated insertional mutagenesis. In this review, we present the biology of the RAG genes, the challenges faced during the development of viral gene addition for RAG1-SCID, and the current status of gene therapy for RAG1 deficiency. In particular, we highlight the latest advances and challenges in CRISPR/Cas9 gene editing and their potential for the future of gene therapy.
Collapse
Affiliation(s)
- Giorgio Gilioli
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arjan C Lankester
- Department of Pediatrics, Pediatric Stem Cell Transplantation Program and Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, the Netherlands
| | - Sander de Kivit
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frank J T Staal
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Lisa M Ott de Bruin
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands; Department of Pediatrics, Pediatric Stem Cell Transplantation Program and Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, the Netherlands
| |
Collapse
|
32
|
Zhu M, Fang Y, Sun Y, Li S, Yu J, Xiong B, Wen C, Zhou B, Huang B, Yin H, Xu H. Sonogenetics in the Treatment of Chronic Diseases: A New Method for Cell Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407373. [PMID: 39488795 PMCID: PMC11672274 DOI: 10.1002/advs.202407373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Sonogenetics is an innovative technology that integrates ultrasound with genetic editing to precisely modulate cellular activities in a non-invasive manner. This method entails introducing and activating mechanosensitive channels on the cell membrane of specific cells using gene delivery vectors. When exposed to ultrasound, these channels can be manipulated to open or close, thereby impacting cellular functions. Sonogenetics is currently being used extensively in the treatment of various chronic diseases, including Parkinson's disease, vision restoration, and cancer therapy. This paper provides a comprehensive review of key components of sonogenetics and focuses on evaluating its prospects and potential challenges in the treatment of chronic disease.
Collapse
Affiliation(s)
- Mingrui Zhu
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yan Fang
- Department of Ultrasound, Huashan HospitalFudan UniversityShanghai200040P. R. China
| | - Yikang Sun
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Shaoyue Li
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for TumorShanghai Tenth People's HospitalUltrasound Research and Education InstituteClinical Research Center for Interventional MedicineSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Jifeng Yu
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Bing Xiong
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Congjian Wen
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Boyang Zhou
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Bin Huang
- Zhejiang HospitalHangzhou310013P. R. China
| | - Haohao Yin
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Huixiong Xu
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| |
Collapse
|
33
|
Inen J, Han CM, Farrel DM, Bilousova G, Kogut I. CIRCLE-Seq for Interrogation of Off-Target Gene Editing. J Vis Exp 2024. [PMID: 39555799 PMCID: PMC11912817 DOI: 10.3791/67069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Circularization for In Vitro Reporting of Cleavage Effects by Sequencing (CIRCLE-seq) is a novel technique developed for the impartial identification of unintended cleavage sites of CRISPR-Cas9 through targeted sequencing of CRISPR-Cas9 cleaved DNA. The protocol involves circularizing genomic DNA (gDNA), which is subsequently treated with the Cas9 protein and a guide RNA (gRNA) of interest. Following treatment, the cleaved DNA is purified and prepared as a library for Illumina sequencing. The sequencing process generates paired-end reads, offering comprehensive data on each cleavage site. CIRCLE-seq provides several advantages over other in vitro methods, including minimal sequencing depth requirements, low background, and high enrichment for Cas9-cleaved gDNA. These advantages enhance sensitivity in identifying both intended and unintended cleavage events. This study provides a comprehensive, step-by-step procedure for examining the off-target activity of CRISPR-Cas9 using CIRCLE-seq. As an example, this protocol is validated by mapping genome-wide unintended cleavage sites of CRISPR-Cas9 during the modification of the AAVS1 locus. The entire CIRCLE-seq process can be completed in two weeks, allowing sufficient time for cell growth, DNA purification, library preparation, and Illumina sequencing. The input of sequencing data into the CIRCLE-seq pipeline facilitates streamlined interpretation and analysis of cleavage sites.
Collapse
Affiliation(s)
- Jeffrey Inen
- Department of Dermatology, University of Colorado School of Medicine, Anschutz Medical Campus; Gates Institute, University of Colorado School of Medicine, Anschutz Medical Campus
| | - Chann Makara Han
- Department of Dermatology, University of Colorado School of Medicine, Anschutz Medical Campus; Gates Institute, University of Colorado School of Medicine, Anschutz Medical Campus
| | - David M Farrel
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Anschutz Medical Campus
| | - Ganna Bilousova
- Department of Dermatology, University of Colorado School of Medicine, Anschutz Medical Campus; Gates Institute, University of Colorado School of Medicine, Anschutz Medical Campus
| | - Igor Kogut
- Department of Dermatology, University of Colorado School of Medicine, Anschutz Medical Campus; Gates Institute, University of Colorado School of Medicine, Anschutz Medical Campus;
| |
Collapse
|
34
|
Gupta AO, Azul M, Bhoopalan SV, Abraham A, Bertaina A, Bidgoli A, Bonfim C, DeZern A, Li J, Louis CU, Purtill D, Ruggeri A, Boelens JJ, Prockop S, Sharma A. International Society for Cell & Gene Therapy Stem Cell Engineering Committee report on the current state of hematopoietic stem and progenitor cell-based genomic therapies and the challenges faced. Cytotherapy 2024; 26:1411-1420. [PMID: 38970612 PMCID: PMC11471386 DOI: 10.1016/j.jcyt.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 07/08/2024]
Abstract
Genetic manipulation of hematopoietic stem cells (HSCs) is being developed as a therapeutic strategy for several inherited disorders. This field is rapidly evolving with several novel tools and techniques being employed to achieve desired genetic changes. While commercial products are now available for sickle cell disease, transfusion-dependent β-thalassemia, metachromatic leukodystrophy and adrenoleukodystrophy, several challenges remain in patient selection, HSC mobilization and collection, genetic manipulation of stem cells, conditioning, hematologic recovery and post-transplant complications, financial issues, equity of access and institutional and global preparedness. In this report, we explore the current state of development of these therapies and provide a comprehensive assessment of the challenges these therapies face as well as potential solutions.
Collapse
Affiliation(s)
- Ashish O Gupta
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Melissa Azul
- Division of Hematology and Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Senthil Velan Bhoopalan
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Allistair Abraham
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Alan Bidgoli
- Division of Blood and Marrow Transplantation, Children's Healthcare of Atlanta, Aflac Blood and Cancer Disorders Center, Emory University, Atlanta, Georgia, USA
| | - Carmem Bonfim
- Pediatric Blood and Marrow Transplantation Division and Pelé Pequeno Príncipe Research Institute, Hospital Pequeno Príncipe, Curitiba, Brazil
| | - Amy DeZern
- Bone Marrow Failure and MDS Program, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Jingjing Li
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Duncan Purtill
- Department of Haematology, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | | | - Jaap Jan Boelens
- Stem Cell Transplantation and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Susan Prockop
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| |
Collapse
|
35
|
Ritter P, Oliveto S, Cordiglieri C, Fasciani A, Di Buduo CA, della Volpe L, Bocconi A, Conci C, Miguel CP, Di Micco R, Balduini A, Raimondi MT, Biffo S. A millifluidic bioreactor allows the long term culture of primary lymphocytes or CD34 + hematopoietic cells while allowing the detection of tumorigenic expansion. Front Bioeng Biotechnol 2024; 12:1388312. [PMID: 39416278 PMCID: PMC11479935 DOI: 10.3389/fbioe.2024.1388312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Long-term culture of primary lymphocytes and hematopoietic stem and progenitor cells (HSPCs) is pivotal to their expansion and study. Furthermore, genetic engineering of the above-mentioned primary human cells has several safety needs, including the requirement of efficient in vitro assays for unwanted tumorigenic events. In this work, we tested and optimized the Miniaturized Optically Accessible Bioreactor (MOAB) platform. The MOAB consists of a millifluidic cell culture device with three optically-accessible culture chambers. Inside the MOAB, we inserted a silk-based framework that resembles some properties of the bone marrow environment and cultivated in this device either CD4+ T lymphocytes isolated from healthy donor buffy coat or cord blood-derived hematopoietic CD34+ cells. A fraction of these cells is viable for up to 3 months. Next, we tested the capability of the MOAB to detect tumorigenic events. Serial dilutions of engineered fluorescent tumor cells were mixed with either CD4+ or CD34+ primary cells, and their growth was followed. By this approach, we successfully detected as little as 100 tumorigenic cells mixed with 100,000 primary cells. We found that non-tumorigenic primary cells colonized the silk environment, whereas tumor cells, after an adaptation phase, expanded and entered the circulation. We conclude that the millifluidic platform allows the detection of rare tumorigenic events in the long-term culture of human cells.
Collapse
Affiliation(s)
- Paolo Ritter
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Stefania Oliveto
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Chiara Cordiglieri
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
| | - Alessandra Fasciani
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
| | | | - Lucrezia della Volpe
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Bocconi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | | | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- University School for Advanced Studies IUSS, Pavia, Italy
| | | | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Stefano Biffo
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
36
|
Ismail AM, Witt E, Bouwman T, Clark W, Yates B, Franco M, Fong S. The longitudinal kinetics of AAV5 vector integration profiles and evaluation of clonal expansion in mice. Mol Ther Methods Clin Dev 2024; 32:101294. [PMID: 39104575 PMCID: PMC11298592 DOI: 10.1016/j.omtm.2024.101294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/24/2024] [Indexed: 08/07/2024]
Abstract
Adeno-associated virus (AAV)-based vectors are used clinically for gene transfer and persist as extrachromosomal episomes. A small fraction of vector genomes integrate into the host genome, but the theoretical risk of tumorigenesis depends on vector regulatory features. A mouse model was used to investigate integration profiles of an AAV serotype 5 (AAV5) vector produced using Sf and HEK293 cells that mimic key features of valoctocogene roxaparvovec (AAV5-hFVIII-SQ), a gene therapy for severe hemophilia A. The majority (95%) of vector genome reads were derived from episomes, and mean (± standard deviation) integration frequency was 2.70 ± 1.26 and 1.79 ± 0.86 integrations per 1,000 cells for Sf- and HEK293-produced vector. Longitudinal integration analysis suggested integrations occur primarily within 1 week, at low frequency, and their abundance was stable over time. Integration profiles were polyclonal and randomly distributed. No major differences in integration profiles were observed for either vector production platform, and no integrations were associated with clonal expansion. Integrations were enriched near transcription start sites of genes highly expressed in the liver (p = 1 × 10-4) and less enriched for genes of lower expression. We found no evidence of tumorigenesis or fibrosis caused by the vector integrations.
Collapse
Affiliation(s)
| | - Evan Witt
- BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | | | - Wyatt Clark
- BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| | | | - Matteo Franco
- ProtaGene CGT GmbH, Heidelberg 69120, Germany
- ProtaGene Inc., Burlington, MA 01803, USA
| | - Sylvia Fong
- BioMarin Pharmaceutical Inc., Novato, CA 94949, USA
| |
Collapse
|
37
|
Bouziana S, Bouzianas D. The Current Landscape of Secondary Malignancies after CAR T-Cell Therapies: How Could Malignancies Be Prevented? Int J Mol Sci 2024; 25:9518. [PMID: 39273462 PMCID: PMC11395546 DOI: 10.3390/ijms25179518] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have revolutionised the field of haematological malignancies by achieving impressive remission rates in patients with highly refractory haematological malignancies, improving overall survival. To date, six commercial anti-CD19 and anti-BCMA CAR T-cell products have been approved by the Food and Drug Administration (FDA) for the treatment of relapsed/refractory B-cell haematological malignancies and multiple myeloma. The indications for CAR T-cell therapies are gradually expanding, with these therapies being investigated in a variety of diseases, including non-malignant ones. Despite the great success, there are several challenges surrounding CAR T-cell therapies, such as non-durable responses and high-grade toxicities. In addition, a new safety concern was added by the FDA on 28 November 2023 following reports of T-cell malignancies in patients previously treated with either anti-CD19 or anti-BCMA autologous CAR T-cell therapies both in clinical trials and in the real-world setting. Since then, several reports have been published presenting the incidence and analysing the risks of other secondary malignancies after CAR T-cell therapies. In this opinion article, the current landscape of secondary malignancies after CAR T-cell therapies is presented, along with a proposed strategy for future research aiming at potentially diminishing or abrogating the risk of developing secondary malignancies after CAR T-cell therapies.
Collapse
Affiliation(s)
- Stella Bouziana
- Department of Hematology, King’s College Hospital, London SE59RS, UK
| | - Dimitrios Bouzianas
- BReMeL, Biopharmaceutical and Regenerative Medicine Laboratories, 55534 Thessaloniki, Greece;
| |
Collapse
|
38
|
Araujo AE, Bentler M, Perez Garmendia X, Kaleem A, Fabian C, Morgan M, Hacker UT, Büning H. Adeno-Associated Virus Vectors-a Target of Cellular and Humoral Immunity-are Expanding Their Reach Toward Hematopoietic Stem Cell Modification and Immunotherapies. Hum Gene Ther 2024; 35:586-603. [PMID: 39193633 DOI: 10.1089/hum.2024.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024] Open
Abstract
All current market-approved gene therapy medical products for in vivo gene therapy of monogenic diseases rely on adeno-associated virus (AAV) vectors. Advances in gene editing technologies and vector engineering have expanded the spectrum of target cells and, thus, diseases that can be addressed. Consequently, AAV vectors are now being explored to modify cells of the hematopoietic system, including hematopoietic stem and progenitor cells (HSPCs), to develop novel strategies to treat monogenic diseases, but also to generate cell- and vaccine-based immunotherapies. However, the cell types that represent important new targets for the AAV vector system are centrally involved in immune responses against the vector and its transgene product as discussed briefly in the first part of this review. In the second part, studies exploring AAV vectors for genetic engineering of HSPCs, T and B lymphocytes, and beyond are presented.
Collapse
Affiliation(s)
- Angela E Araujo
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Martin Bentler
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Asma Kaleem
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Claire Fabian
- Laboratory for Vector based immunotherapy, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Medical Department II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Cancer Center Central Germany, Leipzig, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Ulrich T Hacker
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Laboratory for Vector based immunotherapy, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Medical Department II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Cancer Center Central Germany, Leipzig, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| |
Collapse
|
39
|
Kachanov A, Kostyusheva A, Brezgin S, Karandashov I, Ponomareva N, Tikhonov A, Lukashev A, Pokrovsky V, Zamyatnin AA, Parodi A, Chulanov V, Kostyushev D. The menace of severe adverse events and deaths associated with viral gene therapy and its potential solution. Med Res Rev 2024; 44:2112-2193. [PMID: 38549260 DOI: 10.1002/med.22036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, in vivo gene replacement therapy has significantly advanced, resulting in market approval of numerous therapeutics predominantly relying on adeno-associated viral vectors (AAV). While viral vectors have undeniably addressed several critical healthcare challenges, their clinical application has unveiled a range of limitations and safety concerns. This review highlights the emerging challenges in the field of gene therapy. At first, we discuss both the role of biological barriers in viral gene therapy with a focus on AAVs, and review current landscape of in vivo human gene therapy. We delineate advantages and disadvantages of AAVs as gene delivery vehicles, mostly from the safety perspective (hepatotoxicity, cardiotoxicity, neurotoxicity, inflammatory responses etc.), and outline the mechanisms of adverse events in response to AAV. Contribution of every aspect of AAV vectors (genomic structure, capsid proteins) and host responses to injected AAV is considered and substantiated by basic, translational and clinical studies. The updated evaluation of recent AAV clinical trials and current medical experience clearly shows the risks of AAVs that sometimes overshadow the hopes for curing a hereditary disease. At last, a set of established and new molecular and nanotechnology tools and approaches are provided as potential solutions for mitigating or eliminating side effects. The increasing number of severe adverse reactions and, sadly deaths, demands decisive actions to resolve the issue of immune responses and extremely high doses of viral vectors used for gene therapy. In response to these challenges, various strategies are under development, including approaches aimed at augmenting characteristics of viral vectors and others focused on creating secure and efficacious non-viral vectors. This comprehensive review offers an overarching perspective on the present state of gene therapy utilizing both viral and non-viral vectors.
Collapse
Affiliation(s)
- Artyom Kachanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Ivan Karandashov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Andrey Tikhonov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Vadim Pokrovsky
- Laboratory of Biochemical Fundamentals of Pharmacology and Cancer Models, Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People's Friendship University, Russia (RUDN University), Moscow, Russia
| | - Andrey A Zamyatnin
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Research, Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Vladimir Chulanov
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Infectious Diseases, Sechenov University, Moscow, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
40
|
Neřoldová M, Stuchlík A. Chemogenetic Tools and their Use in Studies of Neuropsychiatric Disorders. Physiol Res 2024; 73:S449-S470. [PMID: 38957949 PMCID: PMC11412350 DOI: 10.33549/physiolres.935401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Chemogenetics is a newly developed set of tools that allow for selective manipulation of cell activity. They consist of a receptor mutated irresponsive to endogenous ligands and a synthetic ligand that does not interact with the wild-type receptors. Many different types of these receptors and their respective ligands for inhibiting or excitating neuronal subpopulations were designed in the past few decades. It has been mainly the G-protein coupled receptors (GPCRs) selectively responding to clozapine-N-oxide (CNO), namely Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), that have been employed in research. Chemogenetics offers great possibilities since the activity of the receptors is reversible, inducible on demand by the ligand, and non-invasive. Also, specific groups or types of neurons can be selectively manipulated thanks to the delivery by viral vectors. The effect of the chemogenetic receptors on neurons lasts longer, and even chronic activation can be achieved. That can be useful for behavioral testing. The great advantage of chemogenetic tools is especially apparent in research on brain diseases since they can manipulate whole neuronal circuits and connections between different brain areas. Many psychiatric or other brain diseases revolve around the dysfunction of specific brain networks. Therefore, chemogenetics presents a powerful tool for investigating the underlying mechanisms causing the disease and revealing the link between the circuit dysfunction and the behavioral or cognitive symptoms observed in patients. It could also contribute to the development of more effective treatments.
Collapse
Affiliation(s)
- M Neřoldová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic. E-mail:
| | | |
Collapse
|
41
|
Trejo-Villegas OA, Heijink IH, Ávila-Moreno F. Preclinical evidence in the assembly of mammalian SWI/SNF complexes: Epigenetic insights and clinical perspectives in human lung disease therapy. Mol Ther 2024; 32:2470-2488. [PMID: 38910326 PMCID: PMC11405180 DOI: 10.1016/j.ymthe.2024.06.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/18/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
The SWI/SNF complex, also known as the BRG1/BRM-associated factor (BAF) complex, represents a critical regulator of chromatin remodeling mechanisms in mammals. It is alternatively referred to as mSWI/SNF and has been suggested to be imbalanced in human disease compared with human health. Three types of BAF assemblies associated with it have been described, including (1) canonical BAF (cBAF), (2) polybromo-associated BAF (PBAF), and (3) non-canonical BAF (ncBAF) complexes. Each of these BAF assemblies plays a role, either functional or dysfunctional, in governing gene expression patterns, cellular processes, epigenetic mechanisms, and biological processes. Recent evidence increasingly links the dysregulation of mSWI/SNF complexes to various human non-malignant lung chronic disorders and lung malignant diseases. This review aims to provide a comprehensive general state-of-the-art and a profound examination of the current understanding of mSWI/SNF assembly processes, as well as the structural and functional organization of mSWI/SNF complexes and their subunits. In addition, it explores their intricate functional connections with potentially dysregulated transcription factors, placing particular emphasis on molecular and cellular pathogenic processes in lung diseases. These processes are reflected in human epigenome aberrations that impact clinical and therapeutic levels, suggesting novel perspectives on the diagnosis and molecular therapies for human respiratory diseases.
Collapse
Affiliation(s)
- Octavio A Trejo-Villegas
- Lung Diseases and Functional Epigenomics Laboratory (LUDIFE), Biomedicine Research Unit (UBIMED), Facultad de Estudios Superiores-Iztacala (FES-Iztacala), Universidad Nacional Autónoma de México (UNAM), Avenida de los Barrios #1, Colonia Los Reyes Iztacala, Tlalnepantla de Baz, 54090, Estado de México, México
| | - Irene H Heijink
- Departments of Pathology & Medical Biology and Pulmonology, GRIAC Research Institute, University Medical Center Groningen, University of Groningen, 9713 Groningen, the Netherlands
| | - Federico Ávila-Moreno
- Lung Diseases and Functional Epigenomics Laboratory (LUDIFE), Biomedicine Research Unit (UBIMED), Facultad de Estudios Superiores-Iztacala (FES-Iztacala), Universidad Nacional Autónoma de México (UNAM), Avenida de los Barrios #1, Colonia Los Reyes Iztacala, Tlalnepantla de Baz, 54090, Estado de México, México; Research Unit, Instituto Nacional de Enfermedades Respiratorias (INER), Ismael Cosío Villegas, 14080, Ciudad de México, México; Research Tower, Subdirección de Investigación Básica, Instituto Nacional de Cancerología (INCan), 14080, Ciudad de México, México.
| |
Collapse
|
42
|
Zhang M, Hussain A, Hu B, Yang H, Li C, Guo S, Han X, Li B, Dai Y, Cao Y, Chi H, Weng Y, Qin CF, Huang Y. Atavistic strategy for the treatment of hyperuricemia via ionizable liposomal mRNA. Nat Commun 2024; 15:6463. [PMID: 39085241 PMCID: PMC11292028 DOI: 10.1038/s41467-024-50752-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Hyperuricemia is associated with an increased risk of gout, hypertension, diabetes, and cardiovascular diseases. Most mammals maintain normal serum uric acid (SUA) via urate oxidase (Uox), an enzyme that metabolizes poorly-soluble UA to highly-soluble allantoin. In contrast, Uox became a pseudogene in humans and apes over the long course of evolution. Here we demonstrate an atavistic strategy for treating hyperuricemia based on endogenous expression of Uox in hepatocytes mediated by mRNA (mUox) loaded with an ionizable lipid nanoparticle termed iLAND. mUox@iLAND allows effective transfection and protein expression in vitro. A single dose of mUox@iLAND lowers SUA levels for several weeks in two female murine models, including a novel long-lasting model, which is also confirmed by metabolomics analysis. Together with the excellent safety profiles observed in vivo, the proposed mRNA agent demonstrates substantial potential for hyperuricemia therapy and the prevention of associated conditions.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Bo Hu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Chunhui Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Shuai Guo
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Xiaofeng Han
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
- Rigerna Therapeutics Co. Ltd., Beijing, China
| | - Bei Li
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau, SAR, China
| | - Yuhong Cao
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, China
| | - Hang Chi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuhua Weng
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
43
|
Peterman EL, Ploessl DS, Galloway KE. Accelerating Diverse Cell-Based Therapies Through Scalable Design. Annu Rev Chem Biomol Eng 2024; 15:267-292. [PMID: 38594944 DOI: 10.1146/annurev-chembioeng-100722-121610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Augmenting cells with novel, genetically encoded functions will support therapies that expand beyond natural capacity for immune surveillance and tissue regeneration. However, engineering cells at scale with transgenic cargoes remains a challenge in realizing the potential of cell-based therapies. In this review, we introduce a range of applications for engineering primary cells and stem cells for cell-based therapies. We highlight tools and advances that have launched mammalian cell engineering from bioproduction to precision editing of therapeutically relevant cells. Additionally, we examine how transgenesis methods and genetic cargo designs can be tailored for performance. Altogether, we offer a vision for accelerating the translation of innovative cell-based therapies by harnessing diverse cell types, integrating the expanding array of synthetic biology tools, and building cellular tools through advanced genome writing techniques.
Collapse
Affiliation(s)
- Emma L Peterman
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - Deon S Ploessl
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - Kate E Galloway
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| |
Collapse
|
44
|
Metovic J, Li Y, Gong Y, Eichler F. Gene therapy for the leukodystrophies: From preclinical animal studies to clinical trials. Neurotherapeutics 2024; 21:e00443. [PMID: 39276676 PMCID: PMC11418141 DOI: 10.1016/j.neurot.2024.e00443] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024] Open
Abstract
Leukodystrophies are progressive single gene disorders affecting the white matter of the brain. Several gene therapy trials are in progress to address the urgent unmet need for this patient population. We performed a comprehensive literature review of all gene therapy clinical trials listed in www.clinicaltrials.gov through August 2024, and the relevant preclinical studies that enabled clinical translation. Of the approximately 50 leukodystrophies described to date, only eight have existing gene therapy clinical trials: metachromatic leukodystrophy, X-linked adrenoleukodystrophy, globoid cell leukodystrophy, Canavan disease, giant axonal neuropathy, GM2 gangliosidoses, Alexander disease and Pelizaeus-Merzbacher disease. What led to the emergence of gene therapy trials for these specific disorders? What preclinical data or disease context was enabling? For each of these eight disorders, we first describe its pathophysiology and clinical presentation. We discuss the impact of gene therapy delivery route, targeted cell type, delivery modality, dosage, and timing on therapeutic efficacy. We note that use of allogeneic hematopoietic stem cell transplantation in some leukodystrophies allowed for an accelerated path to clinic even in the absence of available animal models. In other leukodystrophies, small and large animal model studies enabled clinical translation of experimental gene therapies. Human clinical trials for the leukodystrophies include ex vivo lentiviral gene delivery, in vivo AAV-mediated gene delivery, and intrathecal antisense oligonucleotide approaches. We outline adverse events associated with each modality focusing specifically on genotoxicity and immunotoxicity. We review monitoring and management of events related to insertional mutagenesis and immune responses. The data presented in this review show that gene therapy, while promising, requires systematic monitoring to account for the precarious disease biology and the adverse events associated with new technology.
Collapse
Affiliation(s)
- Jasna Metovic
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yedda Li
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yi Gong
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
45
|
Mohammadian Gol T, Zahedipour F, Trosien P, Ureña-Bailén G, Kim M, Antony JS, Mezger M. Gene therapy in pediatrics - Clinical studies and approved drugs (as of 2023). Life Sci 2024; 348:122685. [PMID: 38710276 DOI: 10.1016/j.lfs.2024.122685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Gene therapy in pediatrics represents a cutting-edge therapeutic strategy for treating a range of genetic disorders that manifest in childhood. Gene therapy involves the modification or correction of a mutated gene or the introduction of a functional gene into a patient's cells. In general, it is implemented through two main modalities namely ex vivo gene therapy and in vivo gene therapy. Currently, a noteworthy array of gene therapy products has received valid market authorization, with several others in various stages of the approval process. Additionally, a multitude of clinical trials are actively underway, underscoring the dynamic progress within this field. Pediatric genetic disorders in the fields of hematology, oncology, vision and hearing loss, immunodeficiencies, neurological, and metabolic disorders are areas for gene therapy interventions. This review provides a comprehensive overview of the evolution and current progress of gene therapy-based treatments in the clinic for pediatric patients. It navigates the historical milestones of gene therapies, currently approved gene therapy products by the U.S. Food and Drug Administration (FDA) and/or European Medicines Agency (EMA) for children, and the promising future for genetic disorders. By providing a thorough compilation of approved gene therapy drugs and published results of completed or ongoing clinical trials, this review serves as a guide for pediatric clinicians to get a quick overview of the situation of clinical studies and approved gene therapy products as of 2023.
Collapse
Affiliation(s)
- Tahereh Mohammadian Gol
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Fatemeh Zahedipour
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Paul Trosien
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Guillermo Ureña-Bailén
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Miso Kim
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Justin S Antony
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Markus Mezger
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
46
|
Abou-el-Enein M. The Fate(s) of CAR T-Cell Therapy: Navigating the Risks of CAR+ T-Cell Malignancy. Blood Cancer Discov 2024; 5:249-257. [PMID: 38713831 PMCID: PMC11215381 DOI: 10.1158/2643-3230.bcd-23-0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/09/2024] Open
Abstract
The introduction of chimeric antigen receptor (CAR) T-cell therapy represents a landmark advancement in treating resistant forms of cancer such as leukemia, lymphoma, and myeloma. However, concerns about long-term safety have emerged following an FDA investigation into reports of second primary malignancies (SPM) after CAR-T cell treatment. This review offers a thorough examination of how genetically modified T cells might transform into CAR+ SPM. It explores genetic and molecular pathways leading to T-cell lymphomagenesis, the balance between CAR T-cell persistence, stemness, and oncogenic risk, and the trade-off of T-cell exhaustion, which may limit therapy efficacy but potentially reduce lymphomagenesis risk. Significance: An FDA probe into 22 cases of second primary T-cell malignancies following CAR T-cell therapy stresses the need to investigate their origins. Few may arise from preexisting genetic and epigenetic alterations and those introduced during therapeutic engineering. Technological advances, regulatory oversight, and patient monitoring are essential to mitigate potential risks.
Collapse
Affiliation(s)
- Mohamed Abou-el-Enein
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California.
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California and Children’s Hospital of Los Angeles, Los Angeles, California.
- USC/CHLA Cell Therapy Program, University of Southern California and Children’s Hospital of Los Angeles, Los Angeles, California.
| |
Collapse
|
47
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
48
|
Rossi M, Breman E. Engineering strategies to safely drive CAR T-cells into the future. Front Immunol 2024; 15:1411393. [PMID: 38962002 PMCID: PMC11219585 DOI: 10.3389/fimmu.2024.1411393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/27/2024] [Indexed: 07/05/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has proven a breakthrough in cancer treatment in the last decade, giving unprecedented results against hematological malignancies. All approved CAR T-cell products, as well as many being assessed in clinical trials, are generated using viral vectors to deploy the exogenous genetic material into T-cells. Viral vectors have a long-standing clinical history in gene delivery, and thus underwent iterations of optimization to improve their efficiency and safety. Nonetheless, their capacity to integrate semi-randomly into the host genome makes them potentially oncogenic via insertional mutagenesis and dysregulation of key cellular genes. Secondary cancers following CAR T-cell administration appear to be a rare adverse event. However several cases documented in the last few years put the spotlight on this issue, which might have been underestimated so far, given the relatively recent deployment of CAR T-cell therapies. Furthermore, the initial successes obtained in hematological malignancies have not yet been replicated in solid tumors. It is now clear that further enhancements are needed to allow CAR T-cells to increase long-term persistence, overcome exhaustion and cope with the immunosuppressive tumor microenvironment. To this aim, a variety of genomic engineering strategies are under evaluation, most relying on CRISPR/Cas9 or other gene editing technologies. These approaches are liable to introduce unintended, irreversible genomic alterations in the product cells. In the first part of this review, we will discuss the viral and non-viral approaches used for the generation of CAR T-cells, whereas in the second part we will focus on gene editing and non-gene editing T-cell engineering, with particular regard to advantages, limitations, and safety. Finally, we will critically analyze the different gene deployment and genomic engineering combinations, delineating strategies with a superior safety profile for the production of next-generation CAR T-cell.
Collapse
|
49
|
Chen Y, Shokouhi AR, Voelcker NH, Elnathan R. Nanoinjection: A Platform for Innovation in Ex Vivo Cell Engineering. Acc Chem Res 2024; 57:1722-1735. [PMID: 38819691 PMCID: PMC11191407 DOI: 10.1021/acs.accounts.4c00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
In human cells, intracellular access and therapeutic cargo transport, including gene-editing tools (e.g., CRISPR-Cas9 and transposons), nucleic acids (e.g., DNA, mRNA, and siRNA), peptides, and proteins (e.g., enzymes and antibodies), are tightly constrained to ensure healthy cell function and behavior. This principle is exemplified in the delivery mechanisms of chimeric antigen receptor (CAR)-T cells for ex-vivo immunotherapy. In particular, the clinical success of CAR-T cells has established a new standard of care by curing previously incurable blood cancers. The approach involves the delivery, typically via the use of electroporation (EP) and lentivirus, of therapeutic CAR genes into a patient's own T cells, which are then engineered to express CARs that target and combat their blood cancer. But the key difficulty lies in genetically manipulating these cells without causing irreversible damage or loss of function─all the while minimizing complexities of manufacturing, safety concerns, and costs, and ensuring the efficacy of the final CAR-T cell product.Nanoinjection─the process of intracellular delivery using nanoneedles (NNs)─is an emerging physical delivery route that efficiently negotiates the plasma membrane of many cell types, including primary human T cells. It occurs with minimal perturbation, invasiveness, and toxicity, with high efficiency and throughput at high spatial and temporal resolutions. Nanoinjection promises greatly improved delivery of a broad range of therapeutic cargos with little or no damage to those cargos. A nanoinjection platform allows these cargos to function in the intracellular space as desired. The adaptability of nanoinjection platforms is now bringing major advantages in immunomodulation, mechanotransduction, sampling of cell states (nanobiopsy), controlled intracellular interrogation, and the primary focus of this account─intracellular delivery and its applications in ex vivo cell engineering. Mechanical nanoinjection typically exerts direct mechanical force on the cell membrane, offering a straightforward route to improve membrane perturbation by the NNs and subsequent transport of genetic cargo into targeted cell type (adherent or suspension cells). By contrast, electroactive nanoinjection is controlled by coupling NNs with an electric field─a new route for activating electroporation (EP) at the nanoscale─allowing a dramatic reduction of the applied voltage to a cell and so minimizing post-EP damage to cells and cargo, and overcoming many of the limitations of conventional bulk EP. Nanoinjection transcends mere technique; it is an approach to cell engineering ex vivo, offering the potential to endow cells with new, powerful features such as generating chimeric antigen receptor (CAR)-T cells for future CAR-T cell technologies. We first discuss the manufacturing of NN devices (Section 2), then delve into nanoinjection-mediated cell engineering (Section 3), nanoinjection mechanisms and interfacing methodologies (Section 4), and emerging applications in using nanoinjection to create functional CAR-T cells (Section 5).
Collapse
Affiliation(s)
- Yaping Chen
- Oujiang
Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain
Health), Institute of Aging, Key Laboratory of Alzheimer’s
Disease of Zhejiang Province, Zhejiang Provincial Clinical Research
Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ali-Reza Shokouhi
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Nicolas H. Voelcker
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node of the Australian National
Fabrication Facility, 151 Wellington Road, Clayton, VIC 3168, Australia
- Department
of Materials Science and Engineering, Monash
University, 22 Alliance Lane, Clayton, VIC 3168, Australia
| | - Roey Elnathan
- Monash
Institute of Pharmaceutical Sciences, Monash
University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node of the Australian National
Fabrication Facility, 151 Wellington Road, Clayton, VIC 3168, Australia
- School
of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC 3216, Australia
- Institute
for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, VIC 3216, Australia
- The
Institute for Mental and Physical Health and Clinical Translation,
School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC 3216, Australia
| |
Collapse
|
50
|
Sado T, Cart JB, Lee CL. Mechanisms Underlying the Development of Murine T-Cell Lymphoblastic Lymphoma/Leukemia Induced by Total-Body Irradiation. Cancers (Basel) 2024; 16:2224. [PMID: 38927929 PMCID: PMC11201593 DOI: 10.3390/cancers16122224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Exposure to ionizing radiation is associated with an increased risk of hematologic malignancies in myeloid and lymphoid lineages in humans and experimental mice. Given that substantial evidence links radiation exposure with the risk of hematologic malignancies, it is imperative to deeply understand the mechanisms underlying cellular and molecular changes during the latency period between radiation exposure and the emergence of fully transformed malignant cells. One experimental model widely used in the field of radiation and cancer biology to study hematologic malignancies induced by radiation exposure is mouse models of radiation-induced thymic lymphoma. Murine radiation-induced thymic lymphoma is primarily driven by aberrant activation of Notch signaling, which occurs frequently in human precursor T-cell lymphoblastic lymphoma (T-LBL) and T-cell lymphoblastic leukemia (T-ALL). Here, we summarize the literature elucidating cell-autonomous and non-cell-autonomous mechanisms underlying cancer initiation, progression, and malignant transformation in the thymus following total-body irradiation (TBI) in mice.
Collapse
Affiliation(s)
- Toshihiko Sado
- National Institute of Radiological Sciences, Chiba 263-0024, Japan
| | - John B. Cart
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|