473 results on '"Luigi Naldini"'
Search Results
2. A case of T-cell acute lymphoblastic leukemia in retroviral gene therapy for ADA-SCID
- Author
-
Daniela Cesana, Maria Pia Cicalese, Andrea Calabria, Pietro Merli, Roberta Caruso, Monica Volpin, Laura Rudilosso, Maddalena Migliavacca, Federica Barzaghi, Claudia Fossati, Francesco Gazzo, Simone Pizzi, Andrea Ciolfi, Alessandro Bruselles, Francesca Tucci, Giulio Spinozzi, Giulia Pais, Fabrizio Benedicenti, Matteo Barcella, Ivan Merelli, Pierangela Gallina, Stefania Giannelli, Francesca Dionisio, Serena Scala, Miriam Casiraghi, Luisa Strocchio, Luciana Vinti, Lucia Pacillo, Eleonora Draghi, Marcella Cesana, Sara Riccardo, Chiara Colantuono, Emmanuelle Six, Marina Cavazzana, Filippo Carlucci, Manfred Schmidt, Caterina Cancrini, Fabio Ciceri, Luca Vago, Davide Cacchiarelli, Bernhard Gentner, Luigi Naldini, Marco Tartaglia, Eugenio Montini, Franco Locatelli, and Alessandro Aiuti
- Subjects
Science - Abstract
Abstract Hematopoietic stem cell gene therapy (GT) using a γ-retroviral vector (γ-RV) is an effective treatment for Severe Combined Immunodeficiency due to Adenosine Deaminase deficiency. Here, we describe a case of GT-related T-cell acute lymphoblastic leukemia (T-ALL) that developed 4.7 years after treatment. The patient underwent chemotherapy and haploidentical transplantation and is currently in remission. Blast cells contain a single vector insertion activating the LIM-only protein 2 (LMO2) proto-oncogene, confirmed by physical interaction, and low Adenosine Deaminase (ADA) activity resulting from methylation of viral promoter. The insertion is detected years before T-ALL in multiple lineages, suggesting that further hits occurred in a thymic progenitor. Blast cells contain known and novel somatic mutations as well as germline mutations which may have contributed to transformation. Before T-ALL onset, the insertion profile is similar to those of other ADA-deficient patients. The limited incidence of vector-related adverse events in ADA-deficiency compared to other γ-RV GT trials could be explained by differences in transgenes, background disease and patient’s specific factors.
- Published
- 2024
- Full Text
- View/download PDF
3. GP64-pseudotyped lentiviral vectors target liver endothelial cells and correct hemophilia A mice
- Author
-
Michela Milani, Cesare Canepari, Simone Assanelli, Simone Merlin, Ester Borroni, Francesco Starinieri, Mauro Biffi, Fabio Russo, Anna Fabiano, Desirèe Zambroni, Andrea Annoni, Luigi Naldini, Antonia Follenzi, and Alessio Cantore
- Subjects
In Vivo Gene Therapy ,Lentiviral Vectors ,Liver Endothelial Cells ,Hemophilia A ,Envelope Engineering ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Lentiviral vectors (LV) are efficient vehicles for in vivo gene delivery to the liver. LV integration into the chromatin of target cells ensures their transmission upon proliferation, thus allowing potentially life-long gene therapy following a single administration, even to young individuals. The glycoprotein of the vesicular stomatitis virus (VSV.G) is widely used to pseudotype LV, as it confers broad tropism and high stability. The baculovirus-derived GP64 envelope protein has been proposed as an alternative for in vivo liver-directed gene therapy. Here, we perform a detailed comparison of VSV.G- and GP64-pseudotyped LV in vitro and in vivo. We report that VSV.G-LV transduced hepatocytes better than GP64-LV, however the latter showed improved transduction of liver sinusoidal endothelial cells (LSEC). Combining GP64-pseudotyping with the high surface content of the phagocytosis inhibitor CD47 further enhanced LSEC transduction. Coagulation factor VIII (FVIII), the gene mutated in hemophilia A, is naturally expressed by LSEC, thus we exploited GP64-LV to deliver a FVIII transgene under the control of the endogenous FVIII promoter and achieved therapeutic amounts of FVIII and correction of hemophilia A mice.
- Published
- 2024
- Full Text
- View/download PDF
4. Scalable GMP-compliant gene correction of CD4+ T cells with IDLV template functionally validated in vitro and in vivo
- Author
-
Claudia Asperti, Daniele Canarutto, Simona Porcellini, Francesca Sanvito, Francesca Cecere, Valentina Vavassori, Samuele Ferrari, Elisabetta Rovelli, Luisa Albano, Aurelien Jacob, Lucia Sergi Sergi, Elisa Montaldo, Francesca Ferrua, Luis Ignacio González-Granado, Vassilios Lougaris, Raffaele Badolato, Andrea Finocchi, Anna Villa, Marina Radrizzani, and Luigi Naldini
- Subjects
IDLV ,gene editing ,Cas9 ,GMP ,large-scale process ,hyper-IgM1 ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Hyper-IgM1 is a rare X-linked combined immunodeficiency caused by mutations in the CD40 ligand (CD40LG) gene with a median survival of 25 years, potentially treatable with in situ CD4+ T cell gene editing with Cas9 and a one-size-fits-most corrective donor template. Here, starting from our research-grade editing protocol, we pursued the development of a good manufacturing practice (GMP)-compliant, scalable process that allows for correction, selection and expansion of edited cells, using an integrase defective lentiviral vector as donor template. After systematic optimization of reagents and conditions we proved maintenance of stem and central memory phenotypes and expression and function of CD40LG in edited healthy donor and patient cells recapitulating the physiological CD40LG regulation. We then documented the preserved fitness of edited cells by xenotransplantation into immunodeficient mice. Finally, we transitioned to large-scale manufacturing, and developed a panel of quality control assays. Overall, our GMP-compliant process takes long-range gene editing one step closer to clinical application with a reassuring safety profile.
- Published
- 2023
- Full Text
- View/download PDF
5. TIM-3, LAG-3, or 2B4 gene disruptions increase the anti-tumor response of engineered T cells
- Author
-
Beatrice Claudia Cianciotti, Zulma Irene Magnani, Alessia Ugolini, Barbara Camisa, Ivan Merelli, Valentina Vavassori, Alessia Potenza, Antonio Imparato, Francesco Manfredi, Danilo Abbati, Laura Perani, Antonello Spinelli, Eric Shifrut, Fabio Ciceri, Luca Vago, Raffaella Di Micco, Luigi Naldini, Pietro Genovese, Eliana Ruggiero, and Chiara Bonini
- Subjects
TCR - T cell receptor ,adoptive T cell immunotherapy ,inhibitory receptor ,genome editing ,CRISPR/Cas9 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
BackgroundIn adoptive T cell therapy, the long term therapeutic benefits in patients treated with engineered tumor specific T cells are limited by the lack of long term persistence of the infused cellular products and by the immunosuppressive mechanisms active in the tumor microenvironment. Exhausted T cells infiltrating the tumor are characterized by loss of effector functions triggered by multiple inhibitory receptors (IRs). In patients, IR blockade reverts T cell exhaustion but has low selectivity, potentially unleashing autoreactive clones and resulting in clinical autoimmune side effects. Furthermore, loss of long term protective immunity in cell therapy has been ascribed to the effector memory phenotype of the infused cells.MethodsWe simultaneously redirected T cell specificity towards the NY-ESO-1 antigen via TCR gene editing (TCRED) and permanently disrupted LAG3, TIM-3 or 2B4 genes (IRKO) via CRISPR/Cas9 in a protocol to expand early differentiated long-living memory stem T cells. The effector functions of the TCRED-IRKO and IR competent (TCRED-IRCOMP) cells were tested in short-term co-culture assays and under a chronic stimulation setting in vitro. Finally, the therapeutic efficacy of the developed cellular products were evaluated in multiple myeloma xenograft models.ResultsWe show that upon chronic stimulation, TCRED-IRKO cells are superior to TCRED-IRCOMP cells in resisting functional exhaustion through different mechanisms and efficiently eliminate cancer cells upon tumor re-challenge in vivo. Our data indicate that TIM-3 and 2B4-disruption preserve T-cell degranulation capacity, while LAG-3 disruption prevents the upregulation of additional inhibitory receptors in T cells.ConclusionThese results highlight that TIM-3, LAG-3, and 2B4 disruptions increase the therapeutic benefit of tumor specific cellular products and suggest distinct, non-redundant roles for IRs in anti-tumor responses.
- Published
- 2024
- Full Text
- View/download PDF
6. Harnessing lentiviral vectors for in vivo gene therapy of liver metastases
- Author
-
Giovanna Giacca, Luigi Naldini, and Mario Leonardo Squadrito
- Subjects
cancer immunotherapy ,gene therapy ,lentiviral vectors ,liver metastasis ,Medicine (General) ,R5-920 - Published
- 2024
- Full Text
- View/download PDF
7. 639 Intra-tumor delivery of IFN-alpha by Tie-2 transduced monocytes associated with favorable 2-year survival in unmethylated MGMT GBM patients: preliminary results of TEM-GBM phase 1/2a study
- Author
-
Fabio Ciceri, Alessandro Olivi, Valeria Ferla, PAOLO FERROLI, Francesca Farina, Carlo Russo, Bernhard Gentner, Marica Eoli, Elena Anghileri, Matteo Barcella, Valentina Brambilla, Matteo Carrabba, Valeria Cuccarini, Giorgio D’Alessandris, Francesco Di Meco, Filippo Gagliardi, Federico Legnani, Stefania Mazzoleni, Roberto Pallini, Marco Saini, Silvia Snider, Karen Mullen, Luigi Naldini, and Gaetano Finocchiaro
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
8. P1392: INTERIM ANALYSIS OF FIRST IN HUMAN PHASE I-II CLINICAL TRIAL OF EX-VIVO GENE THERAPY FOR HURLER SYNDROME: AN UPDATE AT 3 YEAR FOLLOW-UP
- Author
-
Francesca Tucci, Giulia Consiglieri, Chiara Filisetti, Maurizio De Pellegrin, Renata Mellone, Francesca Fumagalli, Silvia Darin, Marina Sarzana, Stefano Scarparo, Francesca Ciotti, Paolo Silvani, Cristina Baldoli, Silvia Pontesilli, Rossella Parini, Giancarlo la Marca, Fabio Ciceri, Luigi Naldini, Alessandro Aiuti, Bernhard Gentner, and Maria Ester Bernardo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
9. P1405: IN-DEPTH GENOME INTEGRITY EVALUATION AND GMP-COMPLIANT MANUFACTURING OF HDR GENE EDITED CD4+ T CELLS FOR THE TREATMENT OF HYPER IGM 1
- Author
-
Daniele Canarutto, Claudia Asperti, Valentina Vavassori, Simona Porcellini, Elisabetta Rovelli, Marianna Paulis, Samuele Ferrari, Tiziana Plati, Lucia Sergi Sergi, Anna Villa, Marina Radrizzani, and Luigi Naldini
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
10. S255: CELLULAR SENESCENCE AND INFLAMMATORY PROGRAMS ARE UNINTENDED CONSEQUENCES OF CRISPR-CAS9 GENE EDITING IN HEMATOPOIETIC STEM AND PROGENITOR CELLS
- Author
-
Anastasia Conti, Teresa Tavella, Rosaria De Marco, Federico Midena, Samuele Ferrari, Stefano Beretta, Chiara Brombin, Ivan Merelli, Luigi Naldini, and Raffaella DI Micco
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
11. P1373: UNCOVERING UPSIDES AND PITFALLS OF BASE AND PRIME EDITING IN HEMATOPOIETIC STEM CELLS
- Author
-
Martina Fiumara, Samuele Ferrari, Attya Omer-Javed, Stefano Beretta, Luisa Albano, Daniele Canarutto, Angelica Varesi, Chiara Gaddoni, Chiara Brombin, Federica Cugnata, Erika Zonari, Matteo Maria Naldini, Matteo Barcella, Bernhard Gentner, Ivan Merelli, and Luigi Naldini
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
12. Liver-directed lentiviral gene therapy corrects hemophilia A mice and achieves normal-range factor VIII activity in non-human primates
- Author
-
Michela Milani, Cesare Canepari, Tongyao Liu, Mauro Biffi, Fabio Russo, Tiziana Plati, Rosalia Curto, Susannah Patarroyo-White, Douglas Drager, Ilaria Visigalli, Chiara Brombin, Paola Albertini, Antonia Follenzi, Eduard Ayuso, Christian Mueller, Andrea Annoni, Luigi Naldini, and Alessio Cantore
- Subjects
Science - Abstract
“Lentiviral gene therapy to the liver establishes stable long-term normal to supra-normal coagulation factor VIII activity in mouse models of hemophilia A and in non-human primates, representing a potential new treatment option for people with hemophilia A.”.
- Published
- 2022
- Full Text
- View/download PDF
13. A systematic review and meta-analysis of gene therapy with hematopoietic stem and progenitor cells for monogenic disorders
- Author
-
Francesca Tucci, Stefania Galimberti, Luigi Naldini, Maria Grazia Valsecchi, and Alessandro Aiuti
- Subjects
Science - Abstract
Ex-vivo gene therapy with hematopoietic stem and progenitor cells (HSPCs) is a promising treatment for monogenic diseases. Here the authors report a systematic review and meta-analysis of available evidence assessing clinical outcomes of HSPC gene therapy from clinical trials.
- Published
- 2022
- Full Text
- View/download PDF
14. Myeloid cell‐based delivery of IFN‐γ reprograms the leukemia microenvironment and induces anti‐tumoral immune responses
- Author
-
Adele Mucci, Gabriele Antonarelli, Carolina Caserta, Francesco Maria Vittoria, Giacomo Desantis, Riccardo Pagani, Beatrice Greco, Monica Casucci, Giulia Escobar, Laura Passerini, Nico Lachmann, Francesca Sanvito, Matteo Barcella, Ivan Merelli, Luigi Naldini, and Bernhard Gentner
- Subjects
ex vivo gene therapy ,immunotherapy ,interferon‐gamma ,leukemia ,Tie2‐expressing monocytes ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract The immunosuppressive microenvironment surrounding tumor cells represents a key cause of treatment failure. Therefore, immunotherapies aimed at reprogramming the immune system have largely spread in the past years. We employed gene transfer into hematopoietic stem and progenitor cells to selectively express anti‐tumoral cytokines in tumor‐infiltrating monocytes/macrophages. We show that interferon‐γ (IFN‐γ) reduced tumor progression in mouse models of B‐cell acute lymphoblastic leukemia (B‐ALL) and colorectal carcinoma (MC38). Its activity depended on the immune system's capacity to respond to IFN‐γ and drove the counter‐selection of leukemia cells expressing surrogate antigens. Gene‐based IFN‐γ delivery induced antigen presentation in the myeloid compartment and on leukemia cells, leading to a wave of T cell recruitment and activation, with enhanced clonal expansion of cytotoxic CD8+ T lymphocytes. The activity of IFN‐γ was further enhanced by either co‐delivery of tumor necrosis factor‐α (TNF‐α) or by drugs blocking immunosuppressive escape pathways, with the potential to obtain durable responses.
- Published
- 2021
- Full Text
- View/download PDF
15. Modeling, optimization, and comparable efficacy of T cell and hematopoietic stem cell gene editing for treating hyper‐IgM syndrome
- Author
-
Valentina Vavassori, Elisabetta Mercuri, Genni E Marcovecchio, Maria C Castiello, Giulia Schiroli, Luisa Albano, Carrie Margulies, Frank Buquicchio, Elena Fontana, Stefano Beretta, Ivan Merelli, Andrea Cappelleri, Paola MV Rancoita, Vassilios Lougaris, Alessandro Plebani, Maria Kanariou, Arjan Lankester, Francesca Ferrua, Eugenio Scanziani, Cecilia Cotta‐Ramusino, Anna Villa, Luigi Naldini, and Pietro Genovese
- Subjects
CRISPR‐Cas gene editing ,hematopoietic stem cells ,T‐cell therapy ,truncated EGFR ,X‐linked hyper‐IgM Syndrome ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Precise correction of the CD40LG gene in T cells and hematopoietic stem/progenitor cells (HSPC) holds promise for treating X‐linked hyper‐IgM Syndrome (HIGM1), but its actual therapeutic potential remains elusive. Here, we developed a one‐size‐fits‐all editing strategy for effective T‐cell correction, selection, and depletion and investigated the therapeutic potential of T‐cell and HSPC therapies in the HIGM1 mouse model. Edited patients’ derived CD4 T cells restored physiologically regulated CD40L expression and contact‐dependent B‐cell helper function. Adoptive transfer of wild‐type T cells into conditioned HIGM1 mice rescued antigen‐specific IgG responses and protected mice from a disease‐relevant pathogen. We then obtained ~ 25% CD40LG editing in long‐term repopulating human HSPC. Transplanting such proportion of wild‐type HSPC in HIGM1 mice rescued immune functions similarly to T‐cell therapy. Overall, our findings suggest that autologous edited T cells can provide immediate and substantial benefits to HIGM1 patients and position T‐cell ahead of HSPC gene therapy because of easier translation, lower safety concerns and potentially comparable clinical benefits.
- Published
- 2021
- Full Text
- View/download PDF
16. Laboratory-Scale Lentiviral Vector Production and Purification for Enhanced Ex Vivo and In Vivo Genetic Engineering
- Author
-
Monica Soldi, Lucia Sergi Sergi, Giulia Unali, Thomas Kerzel, Ivan Cuccovillo, Paola Capasso, Andrea Annoni, Mauro Biffi, Paola Maria Vittoria Rancoita, Alessio Cantore, Angelo Lombardo, Luigi Naldini, Mario Leonardo Squadrito, and Anna Kajaste-Rudnitski
- Subjects
Lentiviral vectors ,purification process ,manufacturing ,gene therapy ,ex vivo ,in vivo ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Lentiviral vectors (LVs) are increasingly employed in gene and cell therapy. Standard laboratory production of LVs is not easily scalable, and research-grade LVs often contain contaminants that can interfere with downstream applications. Moreover, purified LV production pipelines have been developed mainly for costly, large-scale, clinical-grade settings. Therefore, a standardized and cost-effective process is still needed to obtain efficient, reproducible, and properly executed experimental studies and preclinical development of ex vivo and in vivo gene therapies, as high infectivity and limited adverse reactions are important factors potentially influencing experimental outcomes also in preclinical settings. We describe here an optimized laboratory-scale workflow whereby an LV-containing supernatant is purified and concentrated by sequential chromatographic steps, obtaining biologically active LVs with an infectious titer and specific activity in the order of 109 transducing unit (TU)/mL and 5 × 104 TU/ng of HIV Gag p24, respectively. The purification workflow removes >99% of the starting plasmid, DNA, and protein impurities, resulting in higher gene transfer and editing efficiency in severe combined immunodeficiency (SCID)-repopulating hematopoietic stem and progenitor cells (HSPCs) ex vivo, as well as reduced activation of inflammatory responses ex vivo and in vivo as compared to TU-matched, laboratory-grade vectors. Our results highlight the value of accessible purified LV production for experimental studies and preclinical testing.
- Published
- 2020
- Full Text
- View/download PDF
17. The EHA Research Roadmap: Hematopoietic Stem Cell Gene Therapy
- Author
-
Luigi Naldini, Maria Pia Cicalese, Maria Ester Bernardo, Bernhard Gentner, Michela Gabaldo, Giuliana Ferrari, and Alessandro Aiuti
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
18. Gene Modification and Three‐Dimensional Scaffolds as Novel Tools to Allow the Use of Postnatal Thymic Epithelial Cells for Thymus Regeneration Approaches
- Author
-
Ileana Bortolomai, Monica Sandri, Elena Draghici, Elena Fontana, Elisabetta Campodoni, Genni Enza Marcovecchio, Francesca Ferrua, Laura Perani, Antonello Spinelli, Tamara Canu, Marco Catucci, Tiziano Di Tomaso, Lucia Sergi Sergi, Antonio Esposito, Angelo Lombardo, Luigi Naldini, Anna Tampieri, Georg A. Hollander, Anna Villa, and Marita Bosticardo
- Subjects
3D collagen scaffolds ,Lentiviral vector ,Thymic epithelial cells ,Thymic regeneration ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Abstract Defective functionality of thymic epithelial cells (TECs), due to genetic mutations or injuring causes, results in altered T‐cell development, leading to immunodeficiency or autoimmunity. These defects cannot be corrected by hematopoietic stem cell transplantation (HSCT), and thymus transplantation has not yet been demonstrated to be fully curative. Here, we provide proof of principle of a novel approach toward thymic regeneration, involving the generation of thymic organoids obtained by seeding gene‐modified postnatal murine TECs into three‐dimensional (3D) collagen type I scaffolds mimicking the thymic ultrastructure. To this end, freshly isolated TECs were transduced with a lentiviral vector system, allowing for doxycycline‐induced Oct4 expression. Transient Oct4 expression promoted TECs expansion without drastically changing the cell lineage identity of adult TECs, which retain the expression of important molecules for thymus functionality such as Foxn1, Dll4, Dll1, and AIRE. Oct4‐expressing TECs (iOCT4 TEC) were able to grow into 3D collagen type I scaffolds both in vitro and in vivo, demonstrating that the collagen structure reproduced a 3D environment similar to the thymic extracellular matrix, perfectly recognized by TECs. In vivo results showed that thymic organoids transplanted subcutaneously in athymic nude mice were vascularized but failed to support thymopoiesis because of their limited in vivo persistence. These findings provide evidence that gene modification, in combination with the usage of 3D biomimetic scaffolds, may represent a novel approach allowing the use of postnatal TECs for thymic regeneration. Stem Cells Translational Medicine 2019;8:1107–1122
- Published
- 2019
- Full Text
- View/download PDF
19. Genetic engineering of hematopoiesis: current stage of clinical translation and future perspectives
- Author
-
Luigi Naldini
- Subjects
gene editing ,gene therapy ,hematopoietic stem cells ,lentiviral vectors ,transplantation ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Here I review the scientific background, current stage of development and future perspectives that I foresee in the field of genetic manipulation of hematopoietic stem cells with a special emphasis on clinical applications.
- Published
- 2019
- Full Text
- View/download PDF
20. Multiple Integrated Non-clinical Studies Predict the Safety of Lentivirus-Mediated Gene Therapy for β-Thalassemia
- Author
-
Maria Rosa Lidonnici, Ylenia Paleari, Francesca Tiboni, Giacomo Mandelli, Claudia Rossi, Michela Vezzoli, Annamaria Aprile, Carsten Werner Lederer, Alessandro Ambrosi, Franck Chanut, Francesca Sanvito, Andrea Calabria, Valentina Poletti, Fulvio Mavilio, Eugenio Montini, Luigi Naldini, Patrizia Cristofori, and Giuliana Ferrari
- Subjects
Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Gene therapy clinical trials require rigorous non-clinical studies in the most relevant models to assess the benefit-to-risk ratio. To support the clinical development of gene therapy for β-thalassemia, we performed in vitro and in vivo studies for prediction of safety. First we developed newly GLOBE-derived vectors that were tested for their transcriptional activity and potential interference with the expression of surrounding genes. Because these vectors did not show significant advantages, GLOBE lentiviral vector (LV) was elected for further safety characterization. To support the use of hematopoietic stem cells (HSCs) transduced by GLOBE LV for the treatment of β-thalassemia, we conducted toxicology, tumorigenicity, and biodistribution studies in compliance with the OECD Principles of Good Laboratory Practice. We demonstrated a lack of toxicity and tumorigenic potential associated with GLOBE LV-transduced cells. Vector integration site (IS) studies demonstrated that both murine and human transduced HSCs retain self-renewal capacity and generate new blood cell progeny in the absence of clonal dominance. Moreover, IS analysis showed an absence of enrichment in cancer-related genes, and the genes targeted by GLOBE LV in human HSCs are well known sites of integration, as seen in other lentiviral gene therapy trials, and have not been associated with clonal expansion. Taken together, these integrated studies provide safety data supporting the clinical application of GLOBE-mediated gene therapy for β-thalassemia. Keywords: thalassemia, gene therapy, hematopoiesis, hematopoietic stem cell, preclinical model, safety, lentiviral vector, biodistribution, genotoxicity, integration site
- Published
- 2018
- Full Text
- View/download PDF
21. Interferon gene therapy reprograms the leukemia microenvironment inducing protective immunity to multiple tumor antigens
- Author
-
Giulia Escobar, Luigi Barbarossa, Giulia Barbiera, Margherita Norelli, Marco Genua, Anna Ranghetti, Tiziana Plati, Barbara Camisa, Chiara Brombin, Davide Cittaro, Andrea Annoni, Attilio Bondanza, Renato Ostuni, Bernhard Gentner, and Luigi Naldini
- Subjects
Science - Abstract
An immune suppressive tumor microenvironment (TME) is a limitation for immunotherapy. Here the authors show that, in a B cell acute lymphoblastic leukemia mouse model, gene-based delivery of IFNα reprograms the leukemia-induced immunosuppressive TME into immunostimulatory and enhances T-cell responses.
- Published
- 2018
- Full Text
- View/download PDF
22. Reversible immortalisation enables genetic correction of human muscle progenitors and engineering of next‐generation human artificial chromosomes for Duchenne muscular dystrophy
- Author
-
Sara Benedetti, Narumi Uno, Hidetoshi Hoshiya, Martina Ragazzi, Giulia Ferrari, Yasuhiro Kazuki, Louise Anne Moyle, Rossana Tonlorenzi, Angelo Lombardo, Soraya Chaouch, Vincent Mouly, Marc Moore, Linda Popplewell, Kanako Kazuki, Motonobu Katoh, Luigi Naldini, George Dickson, Graziella Messina, Mitsuo Oshimura, Giulio Cossu, and Francesco Saverio Tedesco
- Subjects
DMD ,gene therapy ,human artificial chromosomes ,human muscle stem/progenitor cells ,immortalisation ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Transferring large or multiple genes into primary human stem/progenitor cells is challenged by restrictions in vector capacity, and this hurdle limits the success of gene therapy. A paradigm is Duchenne muscular dystrophy (DMD), an incurable disorder caused by mutations in the largest human gene: dystrophin. The combination of large‐capacity vectors, such as human artificial chromosomes (HACs), with stem/progenitor cells may overcome this limitation. We previously reported amelioration of the dystrophic phenotype in mice transplanted with murine muscle progenitors containing a HAC with the entire dystrophin locus (DYS‐HAC). However, translation of this strategy to human muscle progenitors requires extension of their proliferative potential to withstand clonal cell expansion after HAC transfer. Here, we show that reversible cell immortalisation mediated by lentivirally delivered excisable hTERT and Bmi1 transgenes extended cell proliferation, enabling transfer of a novel DYS‐HAC into DMD satellite cell‐derived myoblasts and perivascular cell‐derived mesoangioblasts. Genetically corrected cells maintained a stable karyotype, did not undergo tumorigenic transformation and retained their migration ability. Cells remained myogenic in vitro (spontaneously or upon MyoD induction) and engrafted murine skeletal muscle upon transplantation. Finally, we combined the aforementioned functions into a next‐generation HAC capable of delivering reversible immortalisation, complete genetic correction, additional dystrophin expression, inducible differentiation and controllable cell death. This work establishes a novel platform for complex gene transfer into clinically relevant human muscle progenitors for DMD gene therapy.
- Published
- 2017
- Full Text
- View/download PDF
23. Therapeutic gene editing in CD34+ hematopoietic progenitors from Fanconi anemia patients
- Author
-
Begoña Diez, Pietro Genovese, Francisco J Roman‐Rodriguez, Lara Alvarez, Giulia Schiroli, Laura Ugalde, Sandra Rodriguez‐Perales, Julian Sevilla, Cristina Diaz de Heredia, Michael C Holmes, Angelo Lombardo, Luigi Naldini, Juan Antonio Bueren, and Paula Rio
- Subjects
CD34+ cells ,Fanconi anemia ,gene editing ,hematopoietic stem and progenitor cells ,zinc finger nucleases ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Gene targeting constitutes a new step in the development of gene therapy for inherited diseases. Although previous studies have shown the feasibility of editing fibroblasts from Fanconi anemia (FA) patients, here we aimed at conducting therapeutic gene editing in clinically relevant cells, such as hematopoietic stem cells (HSCs). In our first experiments, we showed that zinc finger nuclease (ZFN)‐mediated insertion of a non‐therapeutic EGFP‐reporter donor in the AAVS1 “safe harbor” locus of FA‐A lymphoblastic cell lines (LCLs), indicating that FANCA is not essential for the editing of human cells. When the same approach was conducted with therapeutic FANCA donors, an efficient phenotypic correction of FA‐A LCLs was obtained. Using primary cord blood CD34+ cells from healthy donors, gene targeting was confirmed not only in in vitro cultured cells, but also in hematopoietic precursors responsible for the repopulation of primary and secondary immunodeficient mice. Moreover, when similar experiments were conducted with mobilized peripheral blood CD34+ cells from FA‐A patients, we could demonstrate for the first time that gene targeting in primary hematopoietic precursors from FA patients is feasible and compatible with the phenotypic correction of these clinically relevant cells.
- Published
- 2017
- Full Text
- View/download PDF
24. Genome editing for scalable production of alloantigen‐free lentiviral vectors for in vivo gene therapy
- Author
-
Michela Milani, Andrea Annoni, Sara Bartolaccini, Mauro Biffi, Fabio Russo, Tiziano Di Tomaso, Andrea Raimondi, Johannes Lengler, Michael C Holmes, Friedrich Scheiflinger, Angelo Lombardo, Alessio Cantore, and Luigi Naldini
- Subjects
gene therapy ,hemophilia ,lentiviral vectors ,MHC‐I ,stable producer cell line ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Lentiviral vectors (LV) are powerful and versatile vehicles for gene therapy. However, their complex biological composition challenges large‐scale manufacturing and raises concerns for in vivo applications, because particle components and contaminants may trigger immune responses. Here, we show that producer cell‐derived polymorphic class‐I major histocompatibility complexes (MHC‐I) are incorporated into the LV surface and trigger allogeneic T‐cell responses. By disrupting the beta‐2 microglobulin gene in producer cells, we obtained MHC‐free LV with substantially reduced immunogenicity. We introduce this targeted editing into a novel stable LV packaging cell line, carrying single‐copy inducible vector components, which can be reproducibly converted into high‐yield LV producers upon site‐specific integration of the LV genome of interest. These LV efficiently transfer genes into relevant targets and are more resistant to complement‐mediated inactivation, because of reduced content of the vesicular stomatitis virus envelope glycoprotein G compared to vectors produced by transient transfection. Altogether, these advances support scalable manufacturing of alloantigen‐free LV with higher purity and increased complement resistance that are better suited for in vivo gene therapy.
- Published
- 2017
- Full Text
- View/download PDF
25. Lentiviral vectors escape innate sensing but trigger p53 in human hematopoietic stem and progenitor cells
- Author
-
Francesco Piras, Michela Riba, Carolina Petrillo, Dejan Lazarevic, Ivan Cuccovillo, Sara Bartolaccini, Elia Stupka, Bernhard Gentner, Davide Cittaro, Luigi Naldini, and Anna Kajaste‐Rudnitski
- Subjects
gene therapy ,hematopoietic stem and progenitor cells ,innate sensing ,lentiviral vectors ,p53 signaling ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Clinical application of lentiviral vector (LV)‐based hematopoietic stem and progenitor cells (HSPC) gene therapy is rapidly becoming a reality. Nevertheless, LV‐mediated signaling and its potential functional consequences on HSPC biology remain poorly understood. We unravel here a remarkably limited impact of LV on the HSPC transcriptional landscape. LV escaped innate immune sensing that instead led to robust IFN responses upon transduction with a gamma‐retroviral vector. However, reverse‐transcribed LV DNA did trigger p53 signaling, activated also by non‐integrating Adeno‐associated vector, ultimately leading to lower cell recovery ex vivo and engraftment in vivo. These effects were more pronounced in the short‐term repopulating cells while long‐term HSC frequencies remained unaffected. Blocking LV‐induced signaling partially rescued both apoptosis and engraftment, highlighting a novel strategy to further dampen the impact of ex vivo gene transfer on HSPC. Overall, our results shed light on viral vector sensing in HSPC and provide critical insight for the development of more stealth gene therapy strategies.
- Published
- 2017
- Full Text
- View/download PDF
26. Efficient Ex Vivo Engineering and Expansion of Highly Purified Human Hematopoietic Stem and Progenitor Cell Populations for Gene Therapy
- Author
-
Erika Zonari, Giacomo Desantis, Carolina Petrillo, Francesco E. Boccalatte, Maria Rosa Lidonnici, Anna Kajaste-Rudnitski, Alessandro Aiuti, Giuliana Ferrari, Luigi Naldini, and Bernhard Gentner
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Ex vivo gene therapy based on CD34+ hematopoietic stem cells (HSCs) has shown promising results in clinical trials, but genetic engineering to high levels and in large scale remains challenging. We devised a sorting strategy that captures more than 90% of HSC activity in less than 10% of mobilized peripheral blood (mPB) CD34+ cells, and modeled a transplantation protocol based on highly purified, genetically engineered HSCs co-infused with uncultured progenitor cells. Prostaglandin E2 stimulation allowed near-complete transduction of HSCs with lentiviral vectors during a culture time of less than 38 hr, mitigating the negative impact of standard culture on progenitor cell function. Exploiting the pyrimidoindole derivative UM171, we show that transduced mPB CD34+CD38− cells with repopulating potential could be expanded ex vivo. Implementing these findings in clinical gene therapy protocols will improve the efficacy, safety, and sustainability of gene therapy and generate new opportunities in the field of gene editing. : In this article, Gentner and colleagues undertake a comprehensive strategy to advance ex vivo genetic engineering of HSCs for gene therapy. They experimentally define an optimal strategy to purify HSCs, which allows uncoupling long-term from short-term hematopoietic reconstitution, and implement ex vivo conditions that best preserve their biological properties applying novel transduction-enhancing compounds and pyrimidoindole derivatives to support HSC expansion. Keywords: HSC gene therapy, purified HSCs, HSC expansion, lentiviral vector transduction, prostaglandin E2, UM171
- Published
- 2017
- Full Text
- View/download PDF
27. Gene therapy for ADA‐SCID, the first marketing approval of an ex vivo gene therapy in Europe: paving the road for the next generation of advanced therapy medicinal products
- Author
-
Alessandro Aiuti, Maria Grazia Roncarolo, and Luigi Naldini
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Gene and cell therapy research recently reached a fundamental milestone toward the goal to deliver new medicines for orphan diseases. In 2016, the European Commission granted market approval to GlaxoSmithKline (GSK) for ex vivo hematopoietic stem cell (HSC) gene therapy for the treatment of adenosine deaminase (ADA)‐deficient severe combined immunodeficiency (SCID), a very rare congenital disorder of the immune system. The new medicine, named Strimvelis™, is an advanced therapy medicinal product (ATMP) (Salmikangas et al, 2015) originally developed by the San Raffaele Telethon Institute for Gene Therapy (SR‐Tiget), a joint venture between Telethon Foundation and San Raffaele Scientific Institute. This ATMP is the first ex vivo stem cell gene therapy to receive regulatory approval anywhere in the world. Strimvelis™ consists of a single infusion of autologous gene‐corrected HSC and is prepared from the patient's own bone marrow (BM) HSCs, which are genetically modified using a gamma‐retroviral vector to insert a functional copy of the ADA gene.
- Published
- 2017
- Full Text
- View/download PDF
28. Pervasive supply of therapeutic lysosomal enzymes in the CNS of normal and Krabbe‐affected non‐human primates by intracerebral lentiviral gene therapy
- Author
-
Vasco Meneghini, Annalisa Lattanzi, Luigi Tiradani, Gabriele Bravo, Francesco Morena, Francesca Sanvito, Andrea Calabria, John Bringas, Jeanne M Fisher‐Perkins, Jason P Dufour, Kate C Baker, Claudio Doglioni, Eugenio Montini, Bruce A Bunnell, Krystof Bankiewicz, Sabata Martino, Luigi Naldini, and Angela Gritti
- Subjects
brain ,gene therapy ,lentiviral vectors ,leukodystrophy ,non‐human primates ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Metachromatic leukodystrophy (MLD) and globoid cell leukodystrophy (GLD or Krabbe disease) are severe neurodegenerative lysosomal storage diseases (LSD) caused by arylsulfatase A (ARSA) and galactosylceramidase (GALC) deficiency, respectively. Our previous studies established lentiviral gene therapy (GT) as a rapid and effective intervention to provide pervasive supply of therapeutic lysosomal enzymes in CNS tissues of MLD and GLD mice. Here, we investigated whether this strategy is similarly effective in juvenile non‐human primates (NHP). To provide proof of principle for tolerability and biological efficacy of the strategy, we established a comprehensive study in normal NHP delivering a clinically relevant lentiviral vector encoding for the human ARSA transgene. Then, we injected a lentiviral vector coding for the human GALC transgene in Krabbe‐affected rhesus macaques, evaluating for the first time the therapeutic potential of lentiviral GT in this unique LSD model. We showed favorable safety profile and consistent pattern of LV transduction and enzyme biodistribution in the two models, supporting the robustness of the proposed GT platform. We documented moderate inflammation at the injection sites, mild immune response to vector particles in few treated animals, no indication of immune response against transgenic products, and no molecular evidence of insertional genotoxicity. Efficient gene transfer in neurons, astrocytes, and oligodendrocytes close to the injection sites resulted in robust production and extensive spreading of transgenic enzymes in the whole CNS and in CSF, leading to supraphysiological ARSA activity in normal NHP and close to physiological GALC activity in the Krabbe NHP, in which biological efficacy was associated with preliminary indication of therapeutic benefit. These results support the rationale for the clinical translation of intracerebral lentiviral GT to address CNS pathology in MLD, GLD, and other neurodegenerative LSD.
- Published
- 2016
- Full Text
- View/download PDF
29. IFNα gene/cell therapy curbs colorectal cancer colonization of the liver by acting on the hepatic microenvironment
- Author
-
Mario Catarinella, Andrea Monestiroli, Giulia Escobar, Amleto Fiocchi, Ngoc Lan Tran, Roberto Aiolfi, Paolo Marra, Antonio Esposito, Federica Cipriani, Luca Aldrighetti, Matteo Iannacone, Luigi Naldini, Luca G Guidotti, and Giovanni Sitia
- Subjects
colorectal cancer ,gene therapy ,interferon‐alpha ,liver metastases ,tumor microenvironment ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Colorectal cancer (CRC) metastatic dissemination to the liver is one of the most life‐threatening malignancies in humans and represents the leading cause of CRC‐related mortality. Herein, we adopted a gene transfer strategy into mouse hematopoietic stem/progenitor cells to generate immune‐competent mice in which TEMs—a subset of Tie2+ monocytes/macrophages found at peritumoral sites—express interferon‐alpha (IFNα), a pleiotropic cytokine with anti‐tumor effects. Utilizing this strategy in mouse models of CRC liver metastasis, we show that TEMs accumulate in the proximity of hepatic metastatic areas and that TEM‐mediated delivery of IFNα inhibits tumor growth when administered prior to metastasis challenge as well as on established hepatic lesions, improving overall survival. Further analyses unveiled that local delivery of IFNα does not inhibit homing but limits the early phases of hepatic CRC cell expansion by acting on the radio‐resistant hepatic microenvironment. TEM‐mediated IFNα expression was not associated with systemic side effects, hematopoietic toxicity, or inability to respond to a virus challenge. Along with the notion that TEMs were detected in the proximity of CRC metastases in human livers, these results raise the possibility to employ similar gene/cell therapies as tumor site‐specific drug‐delivery strategies in patients with CRC.
- Published
- 2016
- Full Text
- View/download PDF
30. Targeted Gene Correction in Osteopetrotic-Induced Pluripotent Stem Cells for the Generation of Functional Osteoclasts
- Author
-
Tui Neri, Sharon Muggeo, Marianna Paulis, Maria Elena Caldana, Laura Crisafulli, Dario Strina, Maria Luisa Focarelli, Francesca Faggioli, Camilla Recordati, Samantha Scaramuzza, Eugenio Scanziani, Stefano Mantero, Chiara Buracchi, Cristina Sobacchi, Angelo Lombardo, Luigi Naldini, Paolo Vezzoni, Anna Villa, and Francesca Ficara
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Autosomal recessive osteopetrosis is a human bone disease mainly caused by TCIRG1 gene mutations that prevent osteoclasts resorbing activity, recapitulated by the oc/oc mouse model. Bone marrow transplantation is the only available treatment, limited by the need for a matched donor. The use of induced pluripotent stem cells (iPSCs) as an unlimited source of autologous cells to generate gene corrected osteoclasts might represent a powerful alternative. We generated iPSCs from oc/oc mice, corrected the mutation using a BAC carrying the entire Tcirg1 gene locus as a template for homologous recombination, and induced hematopoietic differentiation. Similarly to physiologic fetal hematopoiesis, iPSC-derived CD41+ cells gradually gave rise to CD45+ cells, which comprised both mature myeloid cells and high proliferative potential colony-forming cells. Finally, we differentiated the gene corrected iPSC-derived myeloid cells into osteoclasts with rescued bone resorbing activity. These results are promising for a future translation into the human clinical setting.
- Published
- 2015
- Full Text
- View/download PDF
31. Targeted gene therapy and cell reprogramming in Fanconi anemia
- Author
-
Paula Rio, Rocio Baños, Angelo Lombardo, Oscar Quintana‐Bustamante, Lara Alvarez, Zita Garate, Pietro Genovese, Elena Almarza, Antonio Valeri, Begoña Díez, Susana Navarro, Yaima Torres, Juan P Trujillo, Rodolfo Murillas, Jose C Segovia, Enrique Samper, Jordi Surralles, Philip D Gregory, Michael C Holmes, Luigi Naldini, and Juan A Bueren
- Subjects
cell reprogramming ,Fanconi anemia ,gene‐targeting ,iPSCs ,zinc finger nucleases ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Gene targeting is progressively becoming a realistic therapeutic alternative in clinics. It is unknown, however, whether this technology will be suitable for the treatment of DNA repair deficiency syndromes such as Fanconi anemia (FA), with defects in homology‐directed DNA repair. In this study, we used zinc finger nucleases and integrase‐defective lentiviral vectors to demonstrate for the first time that FANCA can be efficiently and specifically targeted into the AAVS1 safe harbor locus in fibroblasts from FA‐A patients. Strikingly, up to 40% of FA fibroblasts showed gene targeting 42 days after gene editing. Given the low number of hematopoietic precursors in the bone marrow of FA patients, gene‐edited FA fibroblasts were then reprogrammed and re‐differentiated toward the hematopoietic lineage. Analyses of gene‐edited FA‐iPSCs confirmed the specific integration of FANCA in the AAVS1 locus in all tested clones. Moreover, the hematopoietic differentiation of these iPSCs efficiently generated disease‐free hematopoietic progenitors. Taken together, our results demonstrate for the first time the feasibility of correcting the phenotype of a DNA repair deficiency syndrome using gene‐targeting and cell reprogramming strategies.
- Published
- 2014
- Full Text
- View/download PDF
32. Liver gene therapy by lentiviral vectors reverses anti‐factor IX pre‐existing immunity in haemophilic mice
- Author
-
Andrea Annoni, Alessio Cantore, Patrizia Della Valle, Kevin Goudy, Mahzad Akbarpour, Fabio Russo, Sara Bartolaccini, Armando D'Angelo, Maria Grazia Roncarolo, and Luigi Naldini
- Subjects
gene therapy ,haemophilia ,immune tolerance ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract A major complication of factor replacement therapy for haemophilia is the development of anti‐factor neutralizing antibodies (inhibitors). Here we show that liver gene therapy by lentiviral vectors (LVs) expressing factor IX (FIX) strongly reduces pre‐existing anti‐FIX antibodies and eradicates FIX inhibitors in haemophilia B mice. Concomitantly, plasma FIX levels and clotting activity rose to 50–100% of normal. The treatment was effective in 75% of treated mice. FIX‐specific plasma cells (PCs) and memory B cells were reduced, likely because of memory B‐cell depletion in response to constant exposure to high doses of FIX. Regulatory T cells displaying FIX‐specific suppressive capacity were induced in gene therapy treated mice and controlled FIX‐specific T helper cells. Gene therapy proved safer than a regimen mimicking immune tolerance induction (ITI) by repeated high‐dose FIX protein administration, which induced severe anaphylactoid reactions in inhibitors‐positive haemophilia B mice. Liver gene therapy can thus reverse pre‐existing immunity, induce active tolerance to FIX and establish sustained FIX activity at therapeutic levels. These data position gene therapy as an attractive treatment option for inhibitors‐positive haemophilic patients.
- Published
- 2013
- Full Text
- View/download PDF
33. TIE2‐expressing monocytes/macrophages regulate revascularization of the ischemic limb
- Author
-
Ashish S. Patel, Alberto Smith, Silvia Nucera, Daniela Biziato, Prakash Saha, Rizwan Q. Attia, Julia Humphries, Katherine Mattock, Steven P. Grover, Oliver T. Lyons, Luca G. Guidotti, Richard Siow, Aleksandar Ivetic, Stuart Egginton, Matthew Waltham, Luigi Naldini, Michele De Palma, and Bijan Modarai
- Subjects
angiogenesis ,limb ischemia ,macrophages ,monocytes ,TIE2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract A third of patients with critical limb ischemia (CLI) will eventually require limb amputation. Therapeutic neovascularization using unselected mononuclear cells to salvage ischemic limbs has produced modest results. The TIE2‐expressing monocytes/macrophages (TEMs) are a myeloid cell subset known to be highly angiogenic in tumours. This study aimed to examine the kinetics of TEMs in patients with CLI and whether these cells promote neovascularization of the ischemic limb. Here we show that there are 10‐fold more circulating TEMs in CLI patients, and removal of ischemia reduces their numbers to normal levels. TEM numbers in ischemic muscle are two‐fold greater than normoxic muscle from the same patient. TEMs from patients with CLI display greater proangiogenic activity than TIE2‐negative monocytes in vitro. Using a mouse model of hindlimb ischemia, lentiviral‐based Tie2 knockdown in TEMs impaired recovery from ischemia, whereas delivery of mouse macrophages overexpressing TIE2, or human TEMs isolated from CLI patients, rescued limb ischemia. These data suggest that enhancing TEM recruitment to the ischemic muscle may have the potential to improve limb neovascularization in CLI patients. →See accompanying articles http://dx.doi.org/10.1002/emmm.201302695 and http://dx.doi.org/10.1002/emmm.201302794
- Published
- 2013
- Full Text
- View/download PDF
34. The first reported generation of several induced pluripotent stem cell lines from homozygous and heterozygous Huntington's disease patients demonstrates mutation related enhanced lysosomal activity
- Author
-
Stefano Camnasio, Alessia Delli Carri, Angelo Lombardo, Iwona Grad, Caterina Mariotti, Alessia Castucci, Björn Rozell, Pietro Lo Riso, Valentina Castiglioni, Chiara Zuccato, Christelle Rochon, Yasuhiro Takashima, Giuseppe Diaferia, Ida Biunno, Cinzia Gellera, Marisa Jaconi, Austin Smith, Outi Hovatta, Luigi Naldini, Stefano Di Donato, Anis Feki, and Elena Cattaneo
- Subjects
Human induced pluripotent stem cells ,Huntington's disease ,Neuronal differentiation ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Neuronal disorders, like Huntington's disease (HD), are difficult to study, due to limited cell accessibility, late onset manifestations, and low availability of material. The establishment of an in vitro model that recapitulates features of the disease may help understanding the cellular and molecular events that trigger disease manifestations. Here, we describe the generation and characterization of a series of induced pluripotent stem (iPS) cells derived from patients with HD, including two rare homozygous genotypes and one heterozygous genotype. We used lentiviral technology to transfer key genes for inducing reprogramming. To confirm pluripotency and differentiation of iPS cells, we used PCR amplification and immunocytochemistry to measure the expression of marker genes in embryoid bodies and neurons. We also analyzed teratomas that formed in iPS cell-injected mice. We found that the length of the pathological CAG repeat did not increase during reprogramming, after long term growth in vitro, and after differentiation into neurons. In addition, we observed no differences between normal and mutant genotypes in reprogramming, growth rate, caspase activation or neuronal differentiation. However, we observed a significant increase in lysosomal activity in HD-iPS cells compared to control iPS cells, both during self-renewal and in iPS-derived neurons.In conclusion, we have established stable HD-iPS cell lines that can be used for investigating disease mechanisms that underlie HD. The CAG stability and lysosomal activity represent novel observations in HD-iPS cells. In the future, these cells may provide the basis for a powerful platform for drug screening and target identification in HD.
- Published
- 2012
- Full Text
- View/download PDF
35. miR-511-3p Modulates Genetic Programs of Tumor-Associated Macrophages
- Author
-
Mario Leonardo Squadrito, Ferdinando Pucci, Laura Magri, Davide Moi, Gregor D. Gilfillan, Anna Ranghetti, Andrea Casazza, Massimiliano Mazzone, Robert Lyle, Luigi Naldini, and Michele De Palma
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Expression of the mannose receptor (MRC1/CD206) identifies macrophage subtypes, such as alternatively activated macrophages (AAMs) and M2-polarized tumor-associated macrophages (TAMs), which are endowed with tissue-remodeling, proangiogenic, and protumoral activity. However, the significance of MRC1 expression for TAM's protumoral activity is unclear. Here, we describe and characterize miR-511-3p, an intronic microRNA (miRNA) encoded by both mouse and human MRC1 genes. By using sensitive miRNA reporter vectors, we demonstrate robust expression and bioactivity of miR-511-3p in MRC1+ AAMs and TAMs. Unexpectedly, enforced expression of miR-511-3p tuned down the protumoral gene signature of MRC1+ TAMs and inhibited tumor growth. Our findings suggest that transcriptional activation of Mrc1 in TAMs evokes a genetic program orchestrated by miR-511-3p, which limits rather than enhances their protumoral functions. Besides uncovering a role for MRC1 as gatekeeper of TAM's protumoral genetic programs, these observations suggest that endogenous miRNAs may operate to establish thresholds for inflammatory cell activation in tumors.
- Published
- 2012
- Full Text
- View/download PDF
36. Monitoring disease evolution and treatment response in lysosomal disorders by the peripheral benzodiazepine receptor ligand PK11195
- Author
-
Ilaria Visigalli, Rosa Maria Moresco, Sara Belloli, Letterio S. Politi, Angela Gritti, Daniela Ungaro, Mario Matarrese, Elia Turolla, Andrea Falini, Giuseppe Scotti, Luigi Naldini, Ferruccio Fazio, and Alessandra Biffi
- Subjects
Lysosomal storage disorders ,Microglia ,Neuroinflammation ,Hematopoietic stem cell transplantation ,Positron emission tomography ,Peripheral benzodiazepine receptor ligands ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Microglia activation and neuroinflammation play a pivotal role in the pathogenesis of lysosomal storage disorders (LSD) affecting the central nervous system (CNS), which are amenable to treatment by hematopoietic stem cell transplantation (HSCT). HSCT efficacy relies on replacing the intra- and extra-vascular hematopoietic cell compartments, including CNS microglia, with a cell population expressing the functional enzyme. Non-invasive and quantitative assessment of microglia activation and of its reduction upon HSCT might allow for evaluation of disease evolution and response to treatment in LSD. We here demonstrate that microglia activation can be quantified ex vivo and in vivo by PET using the peripheral benzodiazepine receptor ligand PK11195 in two models of LSD. Furthermore, we show a differential PBR binding following microglia replacement by donor cells in mice undergoing HSCT. Our data indicates that PBR ligands constitute valuable tools for monitoring the evolution and the response to treatment of LSD with CNS involvement, and enable us to evaluate whether the turnover between endogenous and donor microglia following HSCT could be adequate enough to delay disease progression.
- Published
- 2009
- Full Text
- View/download PDF
37. Design of a regulated lentiviral vector for hematopoietic stem cell gene therapy of globoid cell leukodystrophy
- Author
-
Silvia Ungari, Annita Montepeloso, Francesco Morena, Fabienne Cocchiarella, Alessandra Recchia, Sabata Martino, Bernhard Gentner, Luigi Naldini, and Alessandra Biffi
- Subjects
Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Globoid cell leukodystrophy (GLD) is a demyelinating lysosomal storage disease due to the deficiency of the galactocerebrosidase (GALC) enzyme. The favorable outcome of hematopoietic stem and progenitor cell (HSPC)-based approaches in GLD and other similar diseases suggests HSPC gene therapy as a promising therapeutic option for patients. The path to clinical development of this strategy was hampered by a selective toxicity of the overexpressed GALC in the HSPC compartment. Here, we presented the optimization of a lentiviral vector (LV) in which miR-126 regulation was coupled to codon optimization of the human GALC cDNA to obtain a selective and enhanced enzymatic activity only upon transduced HSPCs differentiation. The safety of human GALC overexpression driven by this LV was extensively demonstrated in vitro and in vivo on human HSPCs from healthy donors. No perturbation in the content of proapoptotic sphingolipids, gene expression profile, and capability of engraftment and mutlilineage differentiation in chimeric mice was observed. The therapeutic potential of this LV was then assessed in a severe GLD murine model that benefited from transplantation of corrected HSPCs with longer survival and ameliorated phenotype as compared to untreated siblings. This construct has thus been selected as a candidate for clinical translation.
- Published
- 2015
- Full Text
- View/download PDF
38. Dynamic Activity of miR-125b and miR-93 during Murine Neural Stem Cell Differentiation In Vitro and in the Subventricular Zone Neurogenic Niche.
- Author
-
Annalisa Lattanzi, Bernhard Gentner, Daniela Corno, Tiziano Di Tomaso, Pieter Mestdagh, Frank Speleman, Luigi Naldini, and Angela Gritti
- Subjects
Medicine ,Science - Abstract
Several microRNAs (miRNAs) that are either specifically enriched or highly expressed in neurons and glia have been described, but the identification of miRNAs modulating neural stem cell (NSC) biology remains elusive. In this study, we exploited high throughput miRNA expression profiling to identify candidate miRNAs enriched in NSC/early progenitors derived from the murine subventricular zone (SVZ). Then, we used lentiviral miRNA sensor vectors (LV.miRT) to monitor the activity of shortlisted miRNAs with cellular and temporal resolution during NSC differentiation, taking advantage of in vitro and in vivo models that recapitulate physiological neurogenesis and gliogenesis and using known neuronal- and glial-specific miRNAs as reference. The LV.miRT platform allowed us monitoring endogenous miRNA activity in low represented cell populations within a bulk culture or within the complexity of CNS tissue, with high sensitivity and specificity. In this way we validated and extended previous results on the neuronal-specific miR-124 and the astroglial-specific miR-23a. Importantly, we describe for the first time a cell type- and differentiation stage-specific modulation of miR-93 and miR-125b in SVZ-derived NSC cultures and in the SVZ neurogenic niche in vivo, suggesting key roles of these miRNAs in regulating NSC function.
- Published
- 2013
- Full Text
- View/download PDF
39. Genetic engineering meets hematopoietic stem cell biology for next-generation gene therapy
- Author
-
Samuele Ferrari, Erika Valeri, Anastasia Conti, Serena Scala, Annamaria Aprile, Raffaella Di Micco, Anna Kajaste-Rudnitski, Eugenio Montini, Giuliana Ferrari, Alessandro Aiuti, and Luigi Naldini
- Subjects
Genetics ,Molecular Medicine ,Cell Biology - Published
- 2023
- Full Text
- View/download PDF
40. Gene-based delivery of immune-activating cytokines for cancer treatment
- Author
-
Federico Rossari, Filippo Birocchi, Luigi Naldini, and Nadia Coltella
- Subjects
Molecular Medicine ,Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
41. Supplementary Methods, Figures 1-3 from Reprogramming T Lymphocytes for Melanoma Adoptive Immunotherapy by T-Cell Receptor Gene Transfer with Lentiviral Vectors
- Author
-
Antonio Rosato, Paola Zanovello, Reno Debets, Ralph A. Willemsen, Luigi Naldini, Mario Amendola, Susanna Mandruzzato, Veronica Tisato, Anna Merlo, Maria Rondina, and Sara Bobisse
- Abstract
Supplementary Methods, Figures 1-3 from Reprogramming T Lymphocytes for Melanoma Adoptive Immunotherapy by T-Cell Receptor Gene Transfer with Lentiviral Vectors
- Published
- 2023
- Full Text
- View/download PDF
42. Supplementary Figures S1 & S2 from MET Overexpression Turns Human Primary Osteoblasts into Osteosarcomas
- Author
-
Maria Flavia Di Renzo, Paolo M. Comoglio, Silvia Giordano, Simona Corso, Riccardo Ferracini, Nicola Baldini, Luigi Naldini, Elisa Vigna, Martina Olivero, Simone Sponza, Barbara Costa, Nadia Coltella, Sofia Avnet, and Salvatore Patanè
- Abstract
Supplementary Figures S1 & S2 from MET Overexpression Turns Human Primary Osteoblasts into Osteosarcomas
- Published
- 2023
- Full Text
- View/download PDF
43. Data from MET Overexpression Turns Human Primary Osteoblasts into Osteosarcomas
- Author
-
Maria Flavia Di Renzo, Paolo M. Comoglio, Silvia Giordano, Simona Corso, Riccardo Ferracini, Nicola Baldini, Luigi Naldini, Elisa Vigna, Martina Olivero, Simone Sponza, Barbara Costa, Nadia Coltella, Sofia Avnet, and Salvatore Patanè
- Abstract
The MET oncogene was causally involved in the pathogenesis of a rare tumor, i.e., the papillary renal cell carcinoma, in which activating mutations, either germline or somatic, were identified. MET activating mutations are rarely found in other human tumors, whereas at higher frequencies, MET is amplified and/or overexpressed in sporadic tumors of specific histotypes, including osteosarcoma. In this work, we provide experimental evidence that overexpression of the MET oncogene causes and sustains the full-blown transformation of osteoblasts. Overexpression of MET, obtained by lentiviral vector–mediated gene transfer, resulted in the conversion of primary human osteoblasts into osteosarcoma cells, displaying the transformed phenotype in vitro and the distinguishing features of human osteosarcomas in vivo. These included atypical nuclei, aberrant mitoses, production of alkaline phosphatase, secretion of osteoid extracellular matrix, and striking neovascularization. Although with a lower tumorigenicity, this phenotype was superimposable to that observed after transfer of the MET gene activated by mutation. Both transformation and tumorigenesis were fully abrogated when MET expression was quenched by short-hairpin RNA or when signaling was impaired by a dominant-negative MET receptor. These data show that MET overexpression is oncogenic and that it is essential for the maintenance of the cancer phenotype. (Cancer Res 2006; 66(9): 4750-7)
- Published
- 2023
- Full Text
- View/download PDF
44. Data from Reprogramming T Lymphocytes for Melanoma Adoptive Immunotherapy by T-Cell Receptor Gene Transfer with Lentiviral Vectors
- Author
-
Antonio Rosato, Paola Zanovello, Reno Debets, Ralph A. Willemsen, Luigi Naldini, Mario Amendola, Susanna Mandruzzato, Veronica Tisato, Anna Merlo, Maria Rondina, and Sara Bobisse
- Abstract
T-cell receptor (TCR) gene transfer for cancer immunotherapy is limited by the availability of large numbers of tumor-specific T cells. TCR α and β chains were isolated from a highly lytic HLA-A2–restricted cytotoxic T lymphocyte (CTL) clone recognizing the melanoma-associated Melan-A/MART-1 antigen and inserted into a lentiviral vector carrying a bidirectional promoter capable of robust and coordinated expression of the two transgenes. Lentiviral vector–based gene delivery systems have shown increased transfer efficiency and transgene expression compared with the widely used γ-retroviral vectors. This vector performed more efficiently than a γ-retrovirus–based vector containing the same expression cassette, resulting in a T-cell population with 60% to 80% of transgenic TCR expression with mainly CD8+ intermediate effector phenotype. Transgenic T cells specifically produced cytokine in response to and killed antigen-expressing melanoma cells, retained an overlapping functional avidity in comparison with the TCR donor CTL clone, and exerted significant therapeutic effects in vivo upon adoptive transfer in melanoma-bearing severe combined immunodeficient mice. Optical imaging showed their accumulation in the tumor site. Overall, our results indicate that lentiviral vectors represent a valid tool for stable and high-intensity expression of transgenic TCR and support clinical exploitation of this approach for therapeutic application. [Cancer Res 2009;69(24):9385–94]
- Published
- 2023
- Full Text
- View/download PDF
45. Supplementary Figure S3 from MET Overexpression Turns Human Primary Osteoblasts into Osteosarcomas
- Author
-
Maria Flavia Di Renzo, Paolo M. Comoglio, Silvia Giordano, Simona Corso, Riccardo Ferracini, Nicola Baldini, Luigi Naldini, Elisa Vigna, Martina Olivero, Simone Sponza, Barbara Costa, Nadia Coltella, Sofia Avnet, and Salvatore Patanè
- Abstract
Supplementary Figure S3 from MET Overexpression Turns Human Primary Osteoblasts into Osteosarcomas
- Published
- 2023
- Full Text
- View/download PDF
46. Hematopoietic Stem- and Progenitor-Cell Gene Therapy for Hurler Syndrome
- Author
-
Bernhard, Gentner, Francesca, Tucci, Stefania, Galimberti, Francesca, Fumagalli, Maurizio, De Pellegrin, Paolo, Silvani, Chiara, Camesasca, Silvia, Pontesilli, Silvia, Darin, Francesca, Ciotti, Marina, Sarzana, Giulia, Consiglieri, Chiara, Filisetti, Giulia, Forni, Laura, Passerini, Daniela, Tomasoni, Daniela, Cesana, Andrea, Calabria, Giulio, Spinozzi, Maria-Pia, Cicalese, Valeria, Calbi, Maddalena, Migliavacca, Federica, Barzaghi, Francesca, Ferrua, Vera, Gallo, Simona, Miglietta, Erika, Zonari, Patali S, Cheruku, Claudia, Forni, Marcella, Facchini, Ambra, Corti, Michela, Gabaldo, Stefano, Zancan, Serena, Gasperini, Attilio, Rovelli, Jaap-Jan, Boelens, Simon A, Jones, Robert, Wynn, Cristina, Baldoli, Eugenio, Montini, Silvia, Gregori, Fabio, Ciceri, Maria G, Valsecchi, Giancarlo, la Marca, Rossella, Parini, Luigi, Naldini, Alessandro, Aiuti, Maria-Ester, Bernardo, Ilaria, Visagalli, Gentner, B, Tucci, F, Galimberti, S, Fumagalli, F, De Pellegrin, M, Silvani, P, Camesasca, C, Pontesilli, S, Darin, S, Ciotti, F, Sarzana, M, Consiglieri, G, Filisetti, C, Forni, G, Passerini, L, Tomasoni, D, Cesana, D, Calabria, A, Spinozzi, G, Cicalese, M, Calbi, V, Migliavacca, M, Barzaghi, F, Ferrua, F, Gallo, V, Miglietta, S, Zonari, E, Cheruku, P, Forni, C, Facchini, M, Corti, A, Gabaldo, M, Zancan, S, Gasperini, S, Rovelli, A, Boelens, J, Jones, S, Wynn, R, Baldoli, C, Montini, E, Gregori, S, Ciceri, F, Valsecchi, M, la Marca, G, Parini, R, Naldini, L, Aiuti, A, Bernardo, M, Gentner, B., Tucci, F., Galimberti, S., Fumagalli, F., De Pellegrin, M., Silvani, P., Camesasca, C., Pontesilli, S., Darin, S., Ciotti, F., Sarzana, M., Consiglieri, G., Filisetti, C., Forni, G., Passerini, L., Tomasoni, D., Cesana, D., Calabria, A., Spinozzi, G., Cicalese, M. -P., Calbi, V., Migliavacca, M., Barzaghi, F., Ferrua, F., Gallo, V., Miglietta, S., Zonari, E., Cheruku, P. S., Forni, C., Facchini, M., Corti, A., Gabaldo, M., Zancan, S., Gasperini, S., Rovelli, A., Boelens, J. -J., Jones, S. A., Wynn, R., Baldoli, C., Montini, E., Gregori, S., Ciceri, F., Valsecchi, M. G., La Marca, G., Parini, R., Naldini, L., Aiuti, A., and Bernardo, M. -E.
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Mucopolysaccharidosis I ,Urinary system ,Genetic enhancement ,Genetic Vectors ,Transplantation, Autologous ,Iduronidase ,Mucopolysaccharidosis type I ,Internal medicine ,MPSIH ,medicine ,Humans ,Progenitor cell ,Hurler syndrome ,Glycosaminoglycans ,business.industry ,Lentivirus ,mucopolysaccharidosis type I ,Hematopoietic Stem Cell Transplantation ,Infant ,Genetic Therapy ,General Medicine ,medicine.disease ,gene therapy ,Transplantation ,Haematopoiesis ,Child, Preschool ,Mutation ,Female ,business ,Ex vivo ,Follow-Up Studies ,Stem Cell Transplantation - Abstract
Background Allogeneic hematopoietic stem-cell transplantation is the standard of care for Hurler syndrome (mucopolysaccharidosis type I, Hurler variant [MPSIH]). However, this treatment is only partially curative and is associated with complications. Methods We are conducting an ongoing study involving eight children with MPSIH. At enrollment, the children lacked a suitable allogeneic donor and had a Developmental Quotient or Intelligence Quotient score above 70 (i.e., none had moderate or severe cognitive impairment). The children received autologous hematopoietic stem and progenitor cells (HSPCs) transduced ex vivo with an alpha-L-iduronidase (IDUA)-encoding lentiviral vector after myeloablative conditioning. Safety and correction of blood IDUA activity up to supraphysiologic levels were the primary end points. Clearance of lysosomal storage material as well as skeletal and neurophysiological development were assessed as secondary and exploratory end points. The planned duration of the study is 5 years. Results We now report interim results. The children's mean (+/- SD) age at the time of HSPC gene therapy was 1.9 +/- 0.5 years. At a median follow-up of 2.10 years, the procedure had a safety profile similar to that known for autologous hematopoietic stem-cell transplantation. All the patients showed prompt and sustained engraftment of gene-corrected cells and had supraphysiologic blood IDUA activity within a month, which was maintained up to the latest follow-up. Urinary glycosaminoglycan (GAG) excretion decreased steeply, reaching normal levels at 12 months in four of five patients who could be evaluated. Previously undetectable levels of IDUA activity in the cerebrospinal fluid became detectable after gene therapy and were associated with local clearance of GAGs. Patients showed stable cognitive performance, stable motor skills corresponding to continued motor development, improved or stable findings on magnetic resonance imaging of the brain and spine, reduced joint stiffness, and normal growth in line with World Health Organization growth charts. Conclusions The delivery of HSPC gene therapy in patients with MPSIH resulted in extensive metabolic correction in peripheral tissues and the central nervous system. (Funded by Fondazione Telethon and others; ClinicalTrials.gov number, ; EudraCT number, .)Hematopoietic Gene Therapy for Hurler Syndrome Eight patients with Hurler syndrome who lacked suitable allogeneic donors received autologous hematopoietic stem and progenitor cells transduced ex vivo with an alpha-L-iduronidase-encoding lentiviral vector. This therapy resulted in extensive metabolic correction in peripheral tissues and the central nervous system.
- Published
- 2021
- Full Text
- View/download PDF
47. Identification of Hepatocyte-Primed Biliary Epithelial Cells in the Homeostatic Liver by in vivo Lentiviral Gene Transfer to Mice and Non-Human Primates
- Author
-
Michela Milani, Francesco Starinieri, Stefano Beretta, Anna Fabiano, Tiziana Plati, Cesare Canepari, Mauro Biffi, Fabio Russo, Rossana Norata, Francesca Sanvito, Ivan Merelli, Luigi Aloia, Meri Huch, Luigi Naldini, and Alessio Cantore
- Published
- 2023
- Full Text
- View/download PDF
48. 648 Autologous macrophage-based immunotherapy Induces a pro-inflammatory state in GBM tumor microenvironment – (TEM-GBM)
- Author
-
Finocchiaro, Gaetano, primary, Gentner, Bernhard, additional, Eoli, Marica, additional, Farina, Francesca, additional, Capotondo, Alessia, additional, Anghileri, Elena, additional, Barcella, Matteo, additional, Brambilla, Valentina, additional, Bruzzone, Maria Grazia, additional, Carrabba, Matteo, additional, Cuccarini, Valeria, additional, D’Alessandris, Giorgio, additional, Meco, Francesco Di, additional, Ferla, Valeria, additional, Franzin, Alberto, additional, Ferroli, Paolo, additional, Gagliardi, Filippo, additional, Legnani, Federico, additional, Mazzoleni, Stefania, additional, Mortini, Pietro, additional, Naldini, Matteo Maria, additional, Olivi, Alessandro, additional, Pallini, Roberto, additional, Patanè, Monica, additional, Paterra, Rosina, additional, Pollo, Bianca, additional, Saini, Marco, additional, Snider, Silvia, additional, Zambanini, Andrew, additional, Luigi, Naldini, additional, Russo, Carlo, additional, and Ciceri, Fabio, additional
- Published
- 2022
- Full Text
- View/download PDF
49. Ensuring a future for gene therapy for rare diseases
- Author
-
Alessandro Aiuti, Francesca Pasinelli, and Luigi Naldini
- Subjects
Rare Diseases ,Humans ,General Medicine ,Genetic Therapy ,General Biochemistry, Genetics and Molecular Biology - Published
- 2022
50. Retrieval of vector integration sites from cell-free DNA
- Author
-
Alessio Cantore, Serena Acquati, Francesca Fumagalli, Fabrizio Benedicenti, Valeria Calbi, Marina Cavazzana, Eugenio Montini, Alessandro Aiuti, Frederic D. Bushman, Pierangela Gallina, Alessandra Magnani, Andrea Calabria, Laura Rudilosso, Maximilian Witzel, Luigi Naldini, Giulio Spinozzi, Christoph Klein, Pietro Genovese, Emmanuelle Six, Alain Fischer, Giulia Schiroli, Daniela Cesana, Cesana, D., Calabria, A., Rudilosso, L., Gallina, P., Benedicenti, F., Spinozzi, G., Schiroli, G., Magnani, A., Acquati, S., Fumagalli, F., Calbi, V., Witzel, M., Bushman, F. D., Cantore, A., Genovese, P., Klein, C., Fischer, A., Cavazzana, M., Six, E., Aiuti, A., Naldini, L., and Montini, E.
- Subjects
0301 basic medicine ,Lymphoma ,Genetic enhancement ,Genetic Vectors ,Computational biology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,law ,In vivo ,medicine ,Humans ,Liquid biopsy ,Polymerase chain reaction ,Leukemia ,Hematopoietic stem cell ,Genetic Therapy ,Leukodystrophy, Metachromatic ,General Medicine ,Genetically modified organism ,030104 developmental biology ,medicine.anatomical_structure ,Cell-free fetal DNA ,chemistry ,030220 oncology & carcinogenesis ,Cell-Free Nucleic Acids ,DNA - Abstract
Gene therapy (GT) has rapidly attracted renewed interest as a treatment for otherwise incurable diseases, with several GT products already on the market and many more entering clinical testing for selected indications. Clonal tracking techniques based on vector integration enable monitoring of the fate of engineered cells in the blood of patients receiving GT and allow assessment of the safety and efficacy of these procedures. However, owing to the limited number of cells that can be tested and the impracticality of studying cells residing in peripheral organs without performing invasive biopsies, this approach provides only a partial snapshot of the clonal repertoire and dynamics of genetically modified cells and reduces the predictive power as a safety readout. In this study, we developed liquid biopsy integration site sequencing, or LiBIS-seq, a polymerase chain reaction technique optimized to quantitatively retrieve vector integration sites from cell-free DNA released into the bloodstream by dying cells residing in several tissues. This approach enabled longitudinal monitoring of in vivo liver-directed GT and clonal tracking in patients receiving hematopoietic stem cell GT, improving our understanding of the clonal composition and turnover of genetically modified cells in solid tissues and, in contrast to conventional analyses based only on circulating blood cells, enabling earlier detection of vector-marked clones that are aberrantly expanding in peripheral tissues.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.