1
|
Borges B, Varthaliti A, Schwab M, Clarke MT, Pivetti C, Gupta N, Cadwell CR, Guibinga G, Phillips S, Del Rio T, Ozsolak F, Imai-Leonard D, Kong L, Laird DJ, Herzeg A, Sumner CJ, MacKenzie TC. Prenatal AAV9-GFP administration in fetal lambs results in transduction of female germ cells and maternal exposure to virus. Mol Ther Methods Clin Dev 2024; 32:101263. [PMID: 38827250 PMCID: PMC11141462 DOI: 10.1016/j.omtm.2024.101263] [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: 01/30/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024]
Abstract
Prenatal somatic cell gene therapy (PSCGT) could potentially treat severe, early-onset genetic disorders such as spinal muscular atrophy (SMA) or muscular dystrophy. Given the approval of adeno-associated virus serotype 9 (AAV9) vectors in infants with SMA by the U.S. Food and Drug Administration, we tested the safety and biodistribution of AAV9-GFP (clinical-grade and dose) in fetal lambs to understand safety and efficacy after umbilical vein or intracranial injection on embryonic day 75 (E75) . Umbilical vein injection led to widespread biodistribution of vector genomes in all examined lamb tissues and in maternal uteruses at harvest (E96 or E140; term = E150). There was robust GFP expression in brain, spinal cord, dorsal root ganglia (DRGs), without DRG toxicity and excellent transduction of diaphragm and quadriceps muscles. However, we found evidence of systemic toxicity (fetal growth restriction) and maternal exposure to the viral vector (transient elevation of total bilirubin and a trend toward elevation in anti-AAV9 antibodies). There were no antibodies against GFP in ewes or lambs. Analysis of fetal gonads demonstrated GFP expression in female (but not male) germ cells, with low levels of integration-specific reads, without integration in select proto-oncogenes. These results suggest potential therapeutic benefit of AAV9 PSCGT for neuromuscular disorders, but warrant caution for exposure of female germ cells.
Collapse
Affiliation(s)
- Beltran Borges
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Antonia Varthaliti
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marisa Schwab
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maria T Clarke
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christopher Pivetti
- Department of Surgery, University of California, Davis, Davis, CA 95817, USA
| | - Nalin Gupta
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics and Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Cathryn R Cadwell
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- Weill Neurohub, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ghiabe Guibinga
- Novartis Institutes for BioMedical Research Biologics Center, San Diego, CA 92121, USA
| | - Shirley Phillips
- Novartis Institutes for BioMedical Research Biologics Center, San Diego, CA 92121, USA
| | - Tony Del Rio
- Novartis Institutes for BioMedical Research Biologics Center, San Diego, CA 92121, USA
| | - Fatih Ozsolak
- Novartis Institutes for BioMedical Research Biologics Center, San Diego, CA 92121, USA
| | - Denise Imai-Leonard
- Comparative Pathology Laboratory, University of California, Davis, Davis, CA 95616, USA
| | - Lingling Kong
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Diana J Laird
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Akos Herzeg
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Tippi C MacKenzie
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Center for Maternal-Fetal Precision Medicine, San Francisco, CA 94158, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics and Benioff Children's Hospital, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
2
|
Lemmens M, Dorsheimer L, Zeller A, Dietz-Baum Y. Non-clinical safety assessment of novel drug modalities: Genome safety perspectives on viral-, nuclease- and nucleotide-based gene therapies. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2024; 896:503767. [PMID: 38821669 DOI: 10.1016/j.mrgentox.2024.503767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/08/2024] [Accepted: 05/13/2024] [Indexed: 06/02/2024]
Abstract
Gene therapies have emerged as promising treatments for various conditions including inherited diseases as well as cancer. Ensuring their safe clinical application requires the development of appropriate safety testing strategies. Several guidelines have been provided by health authorities to address these concerns. These guidelines state that non-clinical testing should be carried out on a case-by-case basis depending on the modality. This review focuses on the genome safety assessment of frequently used gene therapy modalities, namely Adeno Associated Viruses (AAVs), Lentiviruses, designer nucleases and mRNAs. Important safety considerations for these modalities, amongst others, are vector integrations into the patient genome (insertional mutagenesis) and off-target editing. Taking into account the constraints of in vivo studies, health authorities endorse the development of novel approach methodologies (NAMs), which are innovative in vitro strategies for genotoxicity testing. This review provides an overview of NAMs applied to viral and CRISPR/Cas9 safety, including next generation sequencing-based methods for integration site analysis and off-target editing. Additionally, NAMs to evaluate the oncogenicity risk arising from unwanted genomic modifications are discussed. Thus, a range of promising techniques are available to support the safe development of gene therapies. Thorough validation, comparisons and correlations with clinical outcomes are essential to identify the most reliable safety testing strategies. By providing a comprehensive overview of these NAMs, this review aims to contribute to a better understanding of the genome safety perspectives of gene therapies.
Collapse
Affiliation(s)
| | - Lena Dorsheimer
- Research and Development, Preclinical Safety, Sanofi, Industriepark Hoechst, Frankfurt am Main 65926, Germany.
| | - Andreas Zeller
- Pharmaceutical Sciences, pRED Innovation Center Basel, Hoffmann-La Roche Ltd, Basel 4070, Switzerland
| | - Yasmin Dietz-Baum
- Research and Development, Preclinical Safety, Sanofi, Industriepark Hoechst, Frankfurt am Main 65926, Germany
| |
Collapse
|
3
|
Yang S, Xu J, Dai Y, Jin S, Sun Y, Li J, Liu C, Ma X, Chen Z, Chen L, Hou J, Mi JQ, Chen SJ. Neutrophil activation and clonal CAR-T re-expansion underpinning cytokine release syndrome during ciltacabtagene autoleucel therapy in multiple myeloma. Nat Commun 2024; 15:360. [PMID: 38191582 PMCID: PMC10774397 DOI: 10.1038/s41467-023-44648-3] [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: 02/21/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
Cytokine release syndrome (CRS) is the most common complication of chimeric antigen receptor redirected T cells (CAR-T) therapy. CAR-T toxicity management has been greatly improved, but CRS remains a prime safety concern. Here we follow serum cytokine levels and circulating immune cell transcriptomes longitudinally in 26 relapsed/refractory multiple myeloma patients receiving the CAR-T product, ciltacabtagene autoleucel, to understand the immunological kinetics of CRS. We find that although T lymphocytes and monocytes/macrophages are the major overall cytokine source in manifest CRS, neutrophil activation peaks earlier, before the onset of severe symptoms. Intracellularly, signaling activation dominated by JAK/STAT pathway occurred prior to cytokine cascade and displayed regular kinetic changes. CRS severity is accurately described and potentially predicted by temporal cytokine secretion signatures. Notably, CAR-T re-expansion is found in three patients, including a fatal case characterized by somatic TET2-mutation, clonal expanded cytotoxic CAR-T, broadened cytokine profiles and irreversible hepatic toxicity. Together, our findings show that a latent phase with distinct immunological changes precedes manifest CRS, providing an optimal window and potential targets for CRS therapeutic intervention and that CAR-T re-expansion warrants close clinical attention and laboratory investigation to mitigate the lethal risk.
Collapse
Affiliation(s)
- Shuangshuang Yang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shiwei Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Sun
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jianfeng Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenglin Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaolin Ma
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lijuan Chen
- Department of Hematology, First affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Sai-Juan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
4
|
Castiello MC, Di Verniere M, Draghici E, Fontana E, Penna S, Sereni L, Zecchillo A, Minuta D, Uva P, Zahn M, Gil-Farina I, Annoni A, Iaia S, Ott de Bruin LM, Notarangelo LD, Pike-Overzet K, Staal FJT, Villa A, Capo V. Partial correction of immunodeficiency by lentiviral vector gene therapy in mouse models carrying Rag1 hypomorphic mutations. Front Immunol 2023; 14:1268620. [PMID: 38022635 PMCID: PMC10679457 DOI: 10.3389/fimmu.2023.1268620] [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: 07/28/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Recombination activating genes (RAG) 1 and 2 defects are the most frequent form of severe combined immunodeficiency (SCID). Patients with residual RAG activity have a spectrum of clinical manifestations ranging from Omenn syndrome to delayed-onset combined immunodeficiency, often associated with granulomas and/or autoimmunity (CID-G/AI). Lentiviral vector (LV) gene therapy (GT) has been proposed as an alternative treatment to the standard hematopoietic stem cell transplant and a clinical trial for RAG1 SCID patients recently started. However, GT in patients with hypomorphic RAG mutations poses additional risks, because of the residual endogenous RAG1 expression and the general state of immune dysregulation and associated inflammation. Methods In this study, we assessed the efficacy of GT in 2 hypomorphic Rag1 murine models (Rag1F971L/F971L and Rag1R972Q/R972Q), exploiting the same LV used in the clinical trial encoding RAG1 under control of the MND promoter. Results and discussion Starting 6 weeks after transplant, GT-treated mice showed a decrease in proportion of myeloid cells and a concomitant increase of B, T and total white blood cells. However, counts remained lower than in mice transplanted with WT Lin- cells. At euthanasia, we observed a general redistribution of immune subsets in tissues, with the appearance of mature recirculating B cells in the bone marrow. In the thymus, we demonstrated correction of the block at double negative stage, with a modest improvement in the cortical/medullary ratio. Analysis of antigenspecific IgM and IgG serum levels after in vivo challenge showed an amelioration of antibody responses, suggesting that the partial immune correction could confer a clinical benefit. Notably, no overt signs of autoimmunity were detected, with B-cell activating factor decreasing to normal levels and autoantibodies remaining stable after GT. On the other hand, thymic enlargement was frequently observed, although not due to vector integration and insertional mutagenesis. In conclusion, our work shows that GT could partially alleviate the combined immunodeficiency of hypomorphic RAG1 patients and that extensive efficacy and safety studies with alternative models are required before commencing RAG gene therapy in thesehighly complex patients.
Collapse
Affiliation(s)
- Maria Carmina Castiello
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Martina Di Verniere
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Elena Draghici
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elena Fontana
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy
| | - Sara Penna
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Sereni
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Zecchillo
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Denise Minuta
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Uva
- Clinical Bioinformatics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | | | - Andrea Annoni
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Iaia
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lisa M. Ott de Bruin
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Pediatric Stem Cell Transplantation Program, Leiden University Medical Center, Leiden, Netherlands
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Karin Pike-Overzet
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Frank J. T. Staal
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Anna Villa
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Valentina Capo
- San Raffaele-Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| |
Collapse
|
5
|
Banga R, Procopio FA, Lana E, Gladkov GT, Roseto I, Parsons EM, Lian X, Armani-Tourret M, Bellefroid M, Gao C, Kauzlaric A, Foglierini M, Alfageme-Abello O, Sluka SHM, Munoz O, Mastrangelo A, Fenwick C, Muller Y, Mkindi CG, Daubenberger C, Cavassini M, Trunfio R, Déglise S, Corpataux JM, Delorenzi M, Lichterfeld M, Pantaleo G, Perreau M. Lymph node dendritic cells harbor inducible replication-competent HIV despite years of suppressive ART. Cell Host Microbe 2023; 31:1714-1731.e9. [PMID: 37751747 PMCID: PMC11068440 DOI: 10.1016/j.chom.2023.08.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/02/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023]
Abstract
Although gut and lymph node (LN) memory CD4 T cells represent major HIV and simian immunodeficiency virus (SIV) tissue reservoirs, the study of the role of dendritic cells (DCs) in HIV persistence has long been limited to the blood due to difficulties to access lymphoid tissue samples. In this study, we show that LN migratory and resident DC subpopulations harbor distinct phenotypic and transcriptomic profiles. Interestingly, both LN DC subpopulations contain HIV intact provirus and inducible replication-competent HIV despite the expression of the antiviral restriction factor SAMHD1. Notably, LN DC subpopulations isolated from HIV-infected individuals treated for up to 14 years are transcriptionally silent but harbor replication-competent virus that can be induced upon TLR7/8 stimulation. Taken together, these results uncover a potential important contribution of LN DCs to HIV infection in the presence of ART.
Collapse
Affiliation(s)
- Riddhima Banga
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Francesco Andrea Procopio
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Erica Lana
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | | | | | - Elizabeth M Parsons
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Xiaodong Lian
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Annamaria Kauzlaric
- Translational Bioinformatics and Statistics Department of Oncology, University of Lausanne Swiss Cancer Center, Lausanne, Switzerland
| | - Mathilde Foglierini
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Oscar Alfageme-Abello
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Susanna H M Sluka
- Newborn Screening Switzerland, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Olivia Munoz
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Andrea Mastrangelo
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Craig Fenwick
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Yannick Muller
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Catherine Gerald Mkindi
- Ifakara Health Institute, Bagamoyo, United Republic of Tanzania; Department of Medical Parasitology and Infection Biology, Clinical Immunology Unit, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Clinical Immunology Unit, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, 4001 Basel, Switzerland
| | - Matthias Cavassini
- Services of Infectious Diseases, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Rafael Trunfio
- Services of Vascular Surgery, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Sébastien Déglise
- Services of Vascular Surgery, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Jean-Marc Corpataux
- Services of Vascular Surgery, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Mauro Delorenzi
- Translational Bioinformatics and Statistics Department of Oncology, University of Lausanne Swiss Cancer Center, Lausanne, Switzerland
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Giuseppe Pantaleo
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Matthieu Perreau
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland.
| |
Collapse
|
6
|
Retson L, Tiwari N, Vaughn J, Bernes S, Adelson PD, Mansfield K, Libertini S, Kuzmiski B, Alecu I, Gabriel R, Mangum R. Epithelioid neoplasm of the spinal cord in a child with spinal muscular atrophy treated with onasemnogene abeparvovec. Mol Ther 2023; 31:2991-2998. [PMID: 37598295 PMCID: PMC10556221 DOI: 10.1016/j.ymthe.2023.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 08/21/2023] Open
Abstract
Spinal muscular atrophy is an autosomal recessive disease resulting in motor neuron degeneration and progressive life-limiting motor deficits when untreated. Onasemnogene abeparvovec is an adeno-associated virus serotype 9-based gene therapy that improves survival, motor function, and motor milestone achievement in symptomatic and presymptomatic patients. Although the adeno-associated virus genome is maintained as an episome, theoretical risk of tumorigenicity persists should genomic insertion occur. We present the case of a 16-month-old male with spinal muscular atrophy who was diagnosed with an epithelioid neoplasm of the spinal cord approximately 14 months after receiving onasemnogene abeparvovec. In situ hybridization analysis detected an onasemnogene abeparvovec nucleic acid signal broadly distributed in many but not all tumor cells. Integration site analysis on patient formalin-fixed, paraffin-embedded tumor samples failed to detect high-confidence integration sites of onasemnogene abeparvovec. The finding was considered inconclusive because of limited remaining tissue/DNA input. The improved life expectancy resulting from innovative spinal muscular atrophy therapies, including onasemnogene abeparvovec, has created an opportunity to analyze the long-term adverse events and durability of these therapies as well as identify potential disease associations that were previously unrecognized because of the premature death of these patients.
Collapse
Affiliation(s)
- Laura Retson
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Nishant Tiwari
- Pathology & Laboratory Medicine, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Jennifer Vaughn
- Department of Radiology, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Saunder Bernes
- Department of Neurology, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - P David Adelson
- Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Keith Mansfield
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Silvana Libertini
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Brent Kuzmiski
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Iulian Alecu
- Novartis Pharmaceuticals, 4056 Basel, Switzerland
| | | | - Ross Mangum
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ 85016, USA; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA; Creighton University School of Medicine, Phoenix, AZ 85012, USA; Mayo Clinic Alix School of Medicine, Phoenix, AZ 85054, USA.
| |
Collapse
|
7
|
Guiraud J, Le Roy C, Rideau F, Sirand-Pugnet P, Lartigue C, Bébéar C, Arfi Y, Pereyre S. Improved transformation efficiency in Mycoplasma hominis enables disruption of the MIB-MIP system targeting human immunoglobulins. Microbiol Spectr 2023; 11:e0187323. [PMID: 37737635 PMCID: PMC10581049 DOI: 10.1128/spectrum.01873-23] [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: 05/05/2023] [Accepted: 07/18/2023] [Indexed: 09/23/2023] Open
Abstract
The pathogenicity of Mycoplasma hominis is poorly understood, mainly due to the absence of efficient genetic tools. A polyethylene glycol-mediated transformation protocol was recently developed for the M. hominis reference strain M132 using the pMT85-Tet plasmid. The transformation efficiency remained low, hampering generation of a large mutant library. In this study, we improved transformation efficiency by designing M. hominis-specific pMT85 derivatives. Using the Gibson Assembly, the Enterococcus-derived tet(M) gene of the pMT85-Tet plasmid was replaced by that of a M. hominis clinical isolate. Next, the Spiroplasma-derived spiralin gene promoter driving tet(M) expression was substituted by one of three putative regulatory regions (RRs): the M. hominis arginine deiminase RR, the M. hominis elongation factor Tu RR, or the 68 bp SynMyco synthetic RR. SynMyco-based construction led to a 100-fold increase in transformation efficiency in M. hominis M132. This construct was also transformed into the M. hominis PG21 reference strain and three other clinical isolates. The transposon insertion locus was determined for 128 M132-transformants. The majority of the impacted coding sequences encoded lipoproteins and proteins involved in DNA repair or in gene transfer. One transposon integration site was in the mycoplasma immunoglobulin protease gene. Phenotypic characterization of the mutant showed complete disruption of the human antibody cleavage ability of the transformant. These results demonstrate that our M. hominis-optimized plasmid can be used to generate large random transposon insertion libraries, enabling future studies of the pathogenicity of M. hominis. IMPORTANCE Mycoplasma hominis is an opportunistic human pathogen, whose physiopathology is poorly understood and for which genetic tools for transposition mutagenesis have been unavailable for years. A PEG-mediated transformation protocol was developed using the pMT85-Tet plasmid, but the transformation efficiency remained low. We designed a modified pMT85-Tet plasmid suitable for M. hominis. The use of a synthetic regulatory region upstream of the antibiotic resistance marker led to a 100-fold increase in the transformation efficiency. The generation and characterization of large transposon mutagenesis mutant libraries will provide insight into M. hominis pathogenesis. We selected a transformant in which the transposon was integrated in the locus encoding the immunoglobulin cleavage system MIB-MIP. Phenotypic characterization showed that the wild-type strain has a functional MIB-MIP system, whereas the mutant strain had lost the ability to cleave human immunoglobulins.
Collapse
Affiliation(s)
- Jennifer Guiraud
- Centre national de la recherche scientifique (CNRS), UMR 5234 Fundamental Microbiology and Pathogenicity, University of Bordeaux, Bordeaux, France
- Bacteriology Department, National Reference Centre for Bacterial Sexually Transmitted Infections, Bordeaux University Hospital, Bordeaux, France
| | - Chloé Le Roy
- Centre national de la recherche scientifique (CNRS), UMR 5234 Fundamental Microbiology and Pathogenicity, University of Bordeaux, Bordeaux, France
| | - Fabien Rideau
- INRAE, BFP, UMR 1332, Univ. Bordeaux, Villenave d Ornon, France
| | | | - Carole Lartigue
- INRAE, BFP, UMR 1332, Univ. Bordeaux, Villenave d Ornon, France
| | - Cécile Bébéar
- Centre national de la recherche scientifique (CNRS), UMR 5234 Fundamental Microbiology and Pathogenicity, University of Bordeaux, Bordeaux, France
- Bacteriology Department, National Reference Centre for Bacterial Sexually Transmitted Infections, Bordeaux University Hospital, Bordeaux, France
| | - Yonathan Arfi
- INRAE, BFP, UMR 1332, Univ. Bordeaux, Villenave d Ornon, France
| | - Sabine Pereyre
- Centre national de la recherche scientifique (CNRS), UMR 5234 Fundamental Microbiology and Pathogenicity, University of Bordeaux, Bordeaux, France
- Bacteriology Department, National Reference Centre for Bacterial Sexually Transmitted Infections, Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
8
|
Cornetta K, Lin TY, Pellin D, Kohn DB. Meeting FDA Guidance recommendations for replication-competent virus and insertional oncogenesis testing. Mol Ther Methods Clin Dev 2023; 28:28-39. [PMID: 36588821 PMCID: PMC9791246 DOI: 10.1016/j.omtm.2022.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Integrating vectors are associated with alterations in cellular function related to disruption of normal gene function. This has been associated with clonal expansion of cells and, in some instances, cancer. These events have been associated with replication-defective vectors and suggest that the inadvertent exposure to a replication-competent virus arising during vector manufacture would significantly increase the risk of treatment-related adverse events. These risks have led regulatory agencies to require specific monitoring for replication-competent viruses, both prior to and after treatment of patients with gene therapy products. Monitoring the risk of cell expansion and malignancy is also required. In this review, we discuss the rational potential approaches and challenges to meeting the US FDA expectations listed in current guidance documents.
Collapse
Affiliation(s)
- Kenneth Cornetta
- Gene Therapy Testing Laboratory, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- National Gene Vector Biorepository, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tsai-Yu Lin
- Gene Therapy Testing Laboratory, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- National Gene Vector Biorepository, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Danilo Pellin
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Donald B. Kohn
- Departments of Microbiology, Immunology and Molecular Genetics, and Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
9
|
Jain N, Zhao Z, Feucht J, Koche R, Iyer A, Dobrin A, Mansilla-Soto J, Yang J, Zhan Y, Lopez M, Gunset G, Sadelain M. TET2 guards against unchecked BATF3-induced CAR T cell expansion. Nature 2023; 615:315-322. [PMID: 36755094 PMCID: PMC10511001 DOI: 10.1038/s41586-022-05692-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/30/2022] [Indexed: 02/10/2023]
Abstract
Further advances in cell engineering are needed to increase the efficacy of chimeric antigen receptor (CAR) and other T cell-based therapies1-5. As T cell differentiation and functional states are associated with distinct epigenetic profiles6,7, we hypothesized that epigenetic programming may provide a means to improve CAR T cell performance. Targeting the gene that encodes the epigenetic regulator ten-eleven translocation 2 (TET2)8 presents an interesting opportunity as its loss may enhance T cell memory9,10, albeit not cause malignancy9,11,12. Here we show that disruption of TET2 enhances T cell-mediated tumour rejection in leukaemia and prostate cancer models. However, loss of TET2 also enables antigen-independent CAR T cell clonal expansions that may eventually result in prominent systemic tissue infiltration. These clonal proliferations require biallelic TET2 disruption and sustained expression of the AP-1 factor BATF3 to drive a MYC-dependent proliferative program. This proliferative state is associated with reduced effector function that differs from both canonical T cell memory13,14 and exhaustion15,16 states, and is prone to the acquisition of secondary somatic mutations, establishing TET2 as a guardian against BATF3-induced CAR T cell proliferation and ensuing genomic instability. Our findings illustrate the potential of epigenetic programming to enhance T cell immunity but highlight the risk of unleashing unchecked proliferative responses.
Collapse
Affiliation(s)
- Nayan Jain
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Zeguo Zhao
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Judith Feucht
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
- University Children's Hospital, Tübingen, Germany
| | - Richard Koche
- Centre for Epigenetics Research, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Archana Iyer
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Anton Dobrin
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Jorge Mansilla-Soto
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Julie Yang
- Centre for Epigenetics Research, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Yingqian Zhan
- Centre for Epigenetics Research, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Michael Lopez
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Gertrude Gunset
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Michel Sadelain
- Centre for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Centre, New York, NY, USA.
| |
Collapse
|
10
|
Ivančić D, Mir-Pedrol J, Jaraba-Wallace J, Rafel N, Sanchez-Mejias A, Güell M. INSERT-seq enables high-resolution mapping of genomically integrated DNA using Nanopore sequencing. Genome Biol 2022; 23:227. [PMID: 36284361 PMCID: PMC9594898 DOI: 10.1186/s13059-022-02778-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/30/2022] [Indexed: 11/10/2022] Open
Abstract
Comprehensive characterisation of genome engineering technologies is relevant for their development and safe use in human gene therapy. Short-read based methods can overlook insertion events in repetitive regions. We develop INSERT-seq, a method that combines targeted amplification of integrated DNA, UMI-based correction of PCR bias and Oxford Nanopore long-read sequencing for robust analysis of DNA integration. The experimental pipeline improves the number of mappable insertions at repetitive regions by 4.8–7.3% and larger repeats are processed with a computational peak calling pipeline. INSERT-seq is a simple, cheap and robust method to quantitatively characterise DNA integration in diverse ex vivo and in vivo samples.
Collapse
Affiliation(s)
- Dimitrije Ivančić
- grid.5612.00000 0001 2172 2676Departament de Medicina i Ciències de la Vida (MELIS), Universitat Pompeu Fabra, Barcelona, Spain ,grid.473715.30000 0004 6475 7299The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Júlia Mir-Pedrol
- grid.5612.00000 0001 2172 2676Departament de Medicina i Ciències de la Vida (MELIS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Jessica Jaraba-Wallace
- grid.5612.00000 0001 2172 2676Departament de Medicina i Ciències de la Vida (MELIS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Núria Rafel
- grid.5612.00000 0001 2172 2676Departament de Medicina i Ciències de la Vida (MELIS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Avencia Sanchez-Mejias
- grid.5612.00000 0001 2172 2676Departament de Medicina i Ciències de la Vida (MELIS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Marc Güell
- grid.5612.00000 0001 2172 2676Departament de Medicina i Ciències de la Vida (MELIS), Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
11
|
Mesa-Núñez C, Damián C, Fernández-García M, Díez B, Rao G, Schwartz JD, Law KM, Sevilla J, Río P, Yáñez R, Bueren JA, Almarza E. Preclinical safety and efficacy of lentiviral-mediated gene therapy for leukocyte adhesion deficiency type I. MOLECULAR THERAPY - METHODS & CLINICAL DEVELOPMENT 2022; 26:459-470. [PMID: 36092365 PMCID: PMC9418989 DOI: 10.1016/j.omtm.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 07/31/2022] [Indexed: 11/08/2022]
Abstract
Leukocyte adhesion deficiency type I (LAD-I) is a primary immunodeficiency caused by mutations in the ITGB2 gene, which encodes for the CD18 subunit of β2-integrins. Deficient expression of β2-integrins results in impaired neutrophil migration in response to bacterial and fungal infections. Using a lentiviral vector (LV) that mediates a preferential myeloid expression of human CD18 (Chim.hCD18-LV), we first demonstrated that gene therapy efficiently corrected the phenotype of mice with severe LAD-I. Next, we investigated if the ectopic hCD18 expression modified the phenotypic characteristics of human healthy donor hematopoietic stem cells and their progeny. Significantly, transduction of healthy CD34+ cells with the Chim.hCD18-LV did not modify the membrane expression of CD18 nor the adhesion of physiological ligands to transduced cells. Additionally, we observed that the repopulating properties of healthy CD34+ cells were preserved following transduction with the Chim.hCD18-LV, and that a safe polyclonal repopulation pattern was observed in transplanted immunodeficient NOD scid gamma (NSG) mice. In a final set of experiments, we demonstrated that transduction of CD34+ cells from a severe LAD-I patient with the Chim.hCD18-LV restores the expression of β2-integrins in these cells. These results offer additional preclinical safety and efficacy evidence supporting the gene therapy of patients with severe LAD-I.
Collapse
|
12
|
The third-generation anti-CD30 CAR T-cells specifically homing to the tumor and mediating powerful antitumor activity. Sci Rep 2022; 12:10488. [PMID: 35729339 PMCID: PMC9213494 DOI: 10.1038/s41598-022-14523-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 06/08/2022] [Indexed: 12/23/2022] Open
Abstract
CAR T-cell therapy is well tolerated and effective in patients with Hodgkin lymphoma (HL) and anaplastic large cell lymphoma (ALCL). However, even second- generation anti-CD30 CAR T-cells with CD28 (28z) costimulatory domains failed to achieve the desired rate of complete responses. In the present study, we developed second-generation (CD28z) and third-generation (CD28BBz) CAR T-cells targeting CD30 and investigated their efficacy in vitro and in vivo. Both of CD28z and CD28BBz anti-CD30 CAR T cells were similar regarding amplification, T cell subsets distribution, T cell activity, effector/memory and exhaustion. However, we found that the 28BBz anti-CD30 CAR T-cells persist long-term, specifically homing to the tumor and mediating powerful antitumor activity in tumor xenograft models. Subsequently, we also demonstrated that the third generation anti-CD30 CAR T-cells have miner side effects or potential risks of tumorigenesis. Thus, anti-CD30 CAR T-cells represent a safe and effective treatment for Hodgkin lymphoma.
Collapse
|
13
|
Deep Sequencing Analysis of Individual HIV-1 Proviruses Reveals Frequent Asymmetric Long Terminal Repeats. J Virol 2022; 96:e0012222. [PMID: 35674431 PMCID: PMC9278108 DOI: 10.1128/jvi.00122-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Effective strategies to eliminate human immunodeficiency virus type 1 (HIV-1) reservoirs are likely to require more thorough characterizations of proviruses that persist on antiretroviral therapy (ART). The rarity of infected CD4+ T-cells and related technical challenges have limited the characterization of integrated proviruses. Current approaches using next-generation sequencing can be inefficient and limited sequencing depth can make it difficult to link proviral sequences to their respective integration sites. Here, we report on an efficient method by which HIV-1 proviruses and their sites of integration are amplified and sequenced. Across five HIV-1-positive individuals on clinically effective ART, a median of 41.2% (n = 88 of 209) of amplifications yielded near-full-length proviruses and their 5'-host-virus junctions containing a median of 430 bp (range, 18 to 1,363 bp) of flanking host sequence. Unexpectedly, 29.5% (n = 26 of 88) of the sequenced proviruses had structural asymmetries between the 5' and 3' long terminal repeats (LTRs), commonly in the form of major 3' deletions. Sequence-intact proviruses were detected in 3 of 5 donors, and infected CD4+ T-cell clones were detected in 4 of 5 donors. The accuracy of the method was validated by amplifying and sequencing full-length proviruses and flanking host sequences directly from peripheral blood mononuclear cell DNA. The individual proviral sequencing assay (IPSA) described here can provide an accurate, in-depth, and longitudinal characterization of HIV-1 proviruses that persist on ART, which is important for targeting proviruses for elimination and assessing the impact of interventions designed to eradicate HIV-1. IMPORTANCE The integration of human immunodeficiency virus type 1 (HIV-1) into chromosomal DNA establishes the long-term persistence of HIV-1 as proviruses despite effective antiretroviral therapy (ART). Characterizing proviruses is difficult because of their rarity in individuals on long-term suppressive ART, their highly polymorphic sequences and genetic structures, and the need for efficient amplification and sequencing of the provirus and its integration site. Here, we describe a novel, integrated, two-step method (individual proviral sequencing assay [IPSA]) that amplifies the host-virus junction and the full-length provirus except for the last 69 bp of the 3' long terminal repeat (LTR). Using this method, we identified the integration sites of proviruses, including those that are sequence intact and replication competent or defective. Importantly, this new method identified previously unreported asymmetries between LTRs that have implications for how proviruses are detected and quantified. The IPSA method reported is unaffected by LTR asymmetries, permitting a more accurate and comprehensive characterization of the proviral landscape.
Collapse
|
14
|
Wada Y, Sato T, Hasegawa H, Matsudaira T, Nao N, Coler-Reilly ALG, Tasaka T, Yamauchi S, Okagawa T, Momose H, Tanio M, Kuramitsu M, Sasaki D, Matsumoto N, Yagishita N, Yamauchi J, Araya N, Tanabe K, Yamagishi M, Nakashima M, Nakahata S, Iha H, Ogata M, Muramatsu M, Imaizumi Y, Uchimaru K, Miyazaki Y, Konnai S, Yanagihara K, Morishita K, Watanabe T, Yamano Y, Saito M. RAISING is a high-performance method for identifying random transgene integration sites. Commun Biol 2022; 5:535. [PMID: 35654946 PMCID: PMC9163355 DOI: 10.1038/s42003-022-03467-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 05/09/2022] [Indexed: 11/09/2022] Open
Abstract
Both natural viral infections and therapeutic interventions using viral vectors pose significant risks of malignant transformation. Monitoring for clonal expansion of infected cells is important for detecting cancer. Here we developed a novel method of tracking clonality via the detection of transgene integration sites. RAISING (Rapid Amplification of Integration Sites without Interference by Genomic DNA contamination) is a sensitive, inexpensive alternative to established methods. Its compatibility with Sanger sequencing combined with our CLOVA (Clonality Value) software is critical for those without access to expensive high throughput sequencing. We analyzed samples from 688 individuals infected with the retrovirus HTLV-1, which causes adult T-cell leukemia/lymphoma (ATL) to model our method. We defined a clonality value identifying ATL patients with 100% sensitivity and 94.8% specificity, and our longitudinal analysis also demonstrates the usefulness of ATL risk assessment. Future studies will confirm the broad applicability of our technology, especially in the emerging gene therapy sector.
Collapse
Affiliation(s)
- Yusaku Wada
- Biotechnological Research Support Division, FASMAC Co., Ltd, Atsugi, Kanagawa, 243-0021, Japan
| | - Tomoo Sato
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8512, Japan
- Division of Neurology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Hiroo Hasegawa
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8501, Japan
| | - Takahiro Matsudaira
- Biotechnological Research Support Division, FASMAC Co., Ltd, Atsugi, Kanagawa, 243-0021, Japan
| | - Naganori Nao
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- One Health Research Center, Hokkaido University, Sapporo, 060-0818, Japan
| | - Ariella L G Coler-Reilly
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8512, Japan
- Department of Internal Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Shunsuke Yamauchi
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, 060-0818, Japan
| | - Haruka Momose
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Michikazu Tanio
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Madoka Kuramitsu
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Daisuke Sasaki
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
| | - Nariyoshi Matsumoto
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
| | - Naoko Yagishita
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8512, Japan
| | - Junji Yamauchi
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8512, Japan
| | - Natsumi Araya
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8512, Japan
| | - Kenichiro Tanabe
- Pathophysiology and Bioregulation, St. Marianna University Graduate School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Makoto Yamagishi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Makoto Nakashima
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Shingo Nakahata
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Hidekatsu Iha
- Department of Microbiology, Faculty of Medicine, Oita University, Oita, 879-5593, Japan
| | - Masao Ogata
- Department of Hematology, Oita University Hospital, Oita, 879-5593, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Yoshitaka Imaizumi
- Department of Hematology, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
| | - Kaoru Uchimaru
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
- Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8102, Japan
| | - Satoru Konnai
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, 060-0818, Japan
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
- Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8501, Japan
| | - Kazuhiro Morishita
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Toshiki Watanabe
- Department of Practical Management of Medical Information, St. Marianna University Graduate School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Yoshihisa Yamano
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8512, Japan
- Division of Neurology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Masumichi Saito
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.
- Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.
| |
Collapse
|
15
|
Ryu J, Chan W, Wettengel JM, Hanna CB, Burwitz BJ, Hennebold JD, Bimber BN. Rapid, accurate mapping of transgene integration in viable rhesus macaque embryos using enhanced-specificity tagmentation-assisted PCR. Mol Ther Methods Clin Dev 2022; 24:241-254. [PMID: 35211637 PMCID: PMC8829455 DOI: 10.1016/j.omtm.2022.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/16/2022] [Indexed: 11/19/2022]
Abstract
Genome engineering is a powerful tool for in vitro research and the creation of novel model organisms and has growing clinical applications. Randomly integrating vectors, such as lentivirus- or transposase-based methods, are simple and easy to use but carry risks arising from insertional mutagenesis. Here we present enhanced-specificity tagmentation-assisted PCR (esTag-PCR), a rapid and accurate method for mapping transgene integration and copy number. Using stably transfected HepG2 cells, we demonstrate that esTag-PCR has higher integration site detection accuracy and efficiency than alternative tagmentation-based methods. Next, we performed esTag-PCR on rhesus macaque embryos derived from zygotes injected with piggyBac transposase and transposon/transgene plasmid. Using low-input trophectoderm biopsies, we demonstrate that esTag-PCR accurately maps integration events while preserving blastocyst viability. We used these high-resolution data to evaluate the performance of piggyBac-mediated editing of rhesus macaque embryos, demonstrating that increased concentration of transposon/transgene plasmid can increase the fraction of embryos with stable integration; however, the number of integrations per embryo also increases, which may be problematic for some applications. Collectively, esTag-PCR represents an important improvement to the detection of transgene integration, provides a method to validate and screen edited embryos before implantation, and represents an important advance in the creation of transgenic animal models.
Collapse
Affiliation(s)
- Junghyun Ryu
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - William Chan
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jochen M. Wettengel
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, München, 81675 Germany
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Carol B. Hanna
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Benjamin J. Burwitz
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
- Division of Pathobiology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jon D. Hennebold
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Benjamin N. Bimber
- Division of Pathobiology, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
- Corresponding author Benjamin N. Bimber, PhD, Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
16
|
Goyal S, Tisdale J, Schmidt M, Kanter J, Jaroscak J, Whitney D, Bitter H, Gregory PD, Parsons G, Foos M, Yeri A, Gioia M, Voytek SB, Miller A, Lynch J, Colvin RA, Bonner M. Acute Myeloid Leukemia Case after Gene Therapy for Sickle Cell Disease. N Engl J Med 2022; 386:138-147. [PMID: 34898140 DOI: 10.1056/nejmoa2109167] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Gene therapy with LentiGlobin for sickle cell disease (bb1111, lovotibeglogene autotemcel) consists of autologous transplantation of a patient's hematopoietic stem cells transduced with the BB305 lentiviral vector that encodes the βA-T87Q-globin gene. Acute myeloid leukemia developed in a woman approximately 5.5 years after she had received LentiGlobin for sickle cell disease as part of the initial cohort (Group A) of the HGB-206 study. An analysis of peripheral-blood samples revealed that blast cells contained a BB305 lentiviral vector insertion site. The results of an investigation of causality indicated that the leukemia was unlikely to be related to vector insertion, given the location of the insertion site, the very low transgene expression in blast cells, and the lack of an effect on expression of surrounding genes. Several somatic mutations predisposing to acute myeloid leukemia were present after diagnosis, which suggests that patients with sickle cell disease are at increased risk for hematologic malignant conditions after transplantation, most likely because of a combination of risks associated with underlying sickle cell disease, transplantation procedure, and inadequate disease control after treatment. (Funded by Bluebird Bio.).
Collapse
Affiliation(s)
- Sunita Goyal
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - John Tisdale
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Manfred Schmidt
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Julie Kanter
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Jennifer Jaroscak
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Dustin Whitney
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Hans Bitter
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Philip D Gregory
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Geoffrey Parsons
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Marianna Foos
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Ashish Yeri
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Maple Gioia
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Sarah B Voytek
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Alex Miller
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Jessie Lynch
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Richard A Colvin
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| | - Melissa Bonner
- From Bluebird Bio, Cambridge, MA (S.G., D.W., H.B., P.D.G., G.P., M.F., A.Y., M.G., S.B.V., A.M., J.L., R.A.C., M.B.); the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD (J.T.); GeneWerk, Heidelberg, Germany (M.S.); the University of Alabama at Birmingham, Birmingham (J.K.); and the Division of Pediatric Hematology-Oncology, Medical University of South Carolina, Charleston (J.J.)
| |
Collapse
|
17
|
Dastidar S, Majumdar D, Tipanee J, Singh K, Klein AF, Furling D, Chuah MK, VandenDriessche T. Comprehensive transcriptome-wide analysis of spliceopathy correction of myotonic dystrophy using CRISPR-Cas9 in iPSCs-derived cardiomyocytes. Mol Ther 2022; 30:75-91. [PMID: 34371182 PMCID: PMC8753376 DOI: 10.1016/j.ymthe.2021.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 07/01/2021] [Accepted: 07/26/2021] [Indexed: 01/07/2023] Open
Abstract
CTG repeat expansion (CTGexp) is associated with aberrant alternate splicing that contributes to cardiac dysfunction in myotonic dystrophy type 1 (DM1). Excision of this CTGexp repeat using CRISPR-Cas resulted in the disappearance of punctate ribonuclear foci in cardiomyocyte-like cells derived from DM1-induced pluripotent stem cells (iPSCs). This was associated with correction of the underlying spliceopathy as determined by RNA sequencing and alternate splicing analysis. Certain genes were of particular interest due to their role in cardiac development, maturation, and function (TPM4, CYP2J2, DMD, MBNL3, CACNA1H, ROCK2, ACTB) or their association with splicing (SMN2, GCFC2, MBNL3). Moreover, while comparing isogenic CRISPR-Cas9-corrected versus non-corrected DM1 cardiomyocytes, a prominent difference in the splicing pattern for a number of candidate genes was apparent pertaining to genes that are associated with cardiac function (TNNT, TNNT2, TTN, TPM1, SYNE1, CACNA1A, MTMR1, NEBL, TPM1), cellular signaling (NCOR2, CLIP1, LRRFIP2, CLASP1, CAMK2G), and other DM1-related genes (i.e., NUMA1, MBNL2, LDB3) in addition to the disease-causing DMPK gene itself. Subsequent validation using a selected gene subset, including MBNL1, MBNL2, INSR, ADD3, and CRTC2, further confirmed correction of the spliceopathy following CTGexp repeat excision. To our knowledge, the present study provides the first comprehensive unbiased transcriptome-wide analysis of the differential splicing landscape in DM1 patient-derived cardiac cells after excision of the CTGexp repeat using CRISPR-Cas9, showing reversal of the abnormal cardiac spliceopathy in DM1.
Collapse
Affiliation(s)
- Sumitava Dastidar
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Debanjana Majumdar
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Jaitip Tipanee
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Kshitiz Singh
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Arnaud F. Klein
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Denis Furling
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Marinee K. Chuah
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium,Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium,Corresponding author: Marinee K. Chuah, Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| | - Thierry VandenDriessche
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium,Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium,Corresponding author: Thierry VandenDriessche, Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| |
Collapse
|
18
|
Malatinkova E, Thomas J, De Spiegelaere W, Rutsaert S, Geretti AM, Pollakis G, Paxton WA, Vandekerckhove L, Ruggiero A. Measuring Proviral HIV-1 DNA: Hurdles and Improvements to an Assay Monitoring Integration Events Utilising Human Alu Repeat Sequences. Life (Basel) 2021; 11:life11121410. [PMID: 34947941 PMCID: PMC8706387 DOI: 10.3390/life11121410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Integrated HIV-1 DNA persists despite antiretroviral therapy and can fuel viral rebound following treatment interruption. Hence, methods to specifically measure the integrated HIV-1 DNA portion only are important to monitor the reservoir in eradication trials. Here, we provide an up-to-date overview of the literature on the different approaches used to measure integrated HIV-1 DNA. Further, we propose an implemented standard-curve free assay to quantify integrated HIV-1 DNA, so-called Alu-5LTR PCR, which utilises novel primer combinations. We tested the Alu-5LTR PCR in 20 individuals on suppressive ART for a median of nine years; the results were compared to those produced with the standard-free Alu-gag assay. The numbers of median integrated HIV-1 DNA copies were 5 (range: 1–12) and 14 (5–26) with the Alu-gag and Alu-5LTR, respectively. The ratios between Alu-gag vs Alu-5LTR results were distributed within the cohort as follows: most patients (12/20, 60%) provided ratios between 2–5, with 3/20 (15%) and 5/20 (25%) being below or above this range, respectively. Alu-5LTR assay sensitivity was also determined using an “integrated standard”; the data confirmed the increased sensitivity of the assay, i.e., equal to 0.25 proviruses in 10,000 genomes. This work represents an improvement in the field of measuring proviral HIV-1 DNA that could be employed in future HIV-1 persistence and eradication studies.
Collapse
Affiliation(s)
- Eva Malatinkova
- HIV Cure Research Center, Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium; (E.M.); (S.R.); (L.V.)
| | - Jordan Thomas
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (J.T.); (G.P.); (W.A.P.)
| | - Ward De Spiegelaere
- Laboratory of Veterinary Morphology, Faculty of Veterinary Sciences, Ghent University, B-9820 Ghent, Belgium;
| | - Sofie Rutsaert
- HIV Cure Research Center, Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium; (E.M.); (S.R.); (L.V.)
| | - Anna Maria Geretti
- Fondazione PTV and Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
- School of Immunology & Microbial Sciences, King’s College London, London WC2R 2LS, UK
| | - Georgios Pollakis
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (J.T.); (G.P.); (W.A.P.)
| | - William A. Paxton
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (J.T.); (G.P.); (W.A.P.)
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium; (E.M.); (S.R.); (L.V.)
| | - Alessandra Ruggiero
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (J.T.); (G.P.); (W.A.P.)
- Department Neurosciences, Biomedicine and Movement Sciences, School of Medicine-University of Verona, 37129 Verona, Italy
- Correspondence: ; Tel.: +39-045-802-7190
| |
Collapse
|
19
|
Huang AS, Ramos V, Oliveira TY, Gaebler C, Jankovic M, Nussenzweig MC, Cohn LB. Integration features of intact latent HIV-1 in CD4+ T cell clones contribute to viral persistence. J Exp Med 2021; 218:e20211427. [PMID: 34636876 PMCID: PMC8515646 DOI: 10.1084/jem.20211427] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/17/2021] [Accepted: 09/27/2021] [Indexed: 01/26/2023] Open
Abstract
Latent intact HIV-1 proviruses persist in a small subset of long-lived CD4+ T cells that can undergo clonal expansion in vivo. Expanded clones of CD4+ T cells dominate latent reservoirs in individuals on long-term antiretroviral therapy (ART) and represent a major barrier to HIV-1 cure. To determine how integration landscape might contribute to latency, we analyzed integration sites of near full length HIV-1 genomes from individuals on long-term ART, focusing on individuals whose reservoirs are highly clonal. We find that intact proviruses in expanded CD4+ T cell clones are preferentially integrated within Krüppel-associated box (KRAB) domain-containing zinc finger (ZNF) genes. ZNF genes are associated with heterochromatin in memory CD4+ T cells; nevertheless, they are expressed in these cells under steady-state conditions. In contrast to genes carrying unique integrations, ZNF genes carrying clonal intact integrations are down-regulated upon cellular activation. Together, the data suggest selected genomic sites, including ZNF genes, can be especially permissive for maintaining HIV-1 latency during memory CD4+ T cell expansion.
Collapse
Affiliation(s)
- Amy S. Huang
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Victor Ramos
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Thiago Y. Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Christian Gaebler
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Mila Jankovic
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Michel C. Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
- Howard Hughes Medical Institute, Chevy Chase, MD
| | | |
Collapse
|
20
|
Zhang P, Ganesamoorthy D, Nguyen SH, Au R, Coin LJ, Tey SK. Nanopore sequencing as a scalable, cost-effective platform for analyzing polyclonal vector integration sites following clinical T cell therapy. J Immunother Cancer 2021; 8:jitc-2019-000299. [PMID: 32527930 PMCID: PMC7292043 DOI: 10.1136/jitc-2019-000299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2020] [Indexed: 11/21/2022] Open
Abstract
Background Analysis of vector integration sites in gene-modified cells can provide critical information on clonality and potential biological impact on nearby genes. Current short-read next-generation sequencing methods require specialized instruments and large batch runs. Methods We used nanopore sequencing to analyze the vector integration sites of T cells transduced by the gammaretroviral vector, SFG.iCasp9.2A.ΔCD19. DNA from oligoclonal cell lines and polyclonal clinical samples were restriction enzyme digested with two 6-cutters, NcoI and BspHI; and the flanking genomic DNA amplified by inverse PCR or cassette ligation PCR. Following nested PCR and barcoding, the amplicons were sequenced on the Oxford Nanopore platform. Reads were filtered for quality, trimmed, and aligned. Custom tool was developed to cluster reads and merge overlapping clusters. Results Both inverse PCR and cassette ligation PCR could successfully amplify flanking genomic DNA, with cassette ligation PCR showing less bias. The 4.8 million raw reads were grouped into 12,186 clusters and 6410 clones. The 3′long terminal repeat (LTR)-genome junction could be resolved within a 5-nucleotide span for a majority of clusters and within one nucleotide span for clusters with ≥5 reads. The chromosomal distributions of the insertional sites and their predilection for regions proximate to transcription start sites were consistent with previous reports for gammaretroviral vector integrants as analyzed by short-read next-generation sequencing. Conclusion Our study shows that it is feasible to use nanopore sequencing to map polyclonal vector integration sites. The assay is scalable and requires minimum capital, which together enable cost-effective and timely analysis. Further refinement is required to reduce amplification bias and improve single nucleotide resolution.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Devika Ganesamoorthy
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Son Hoang Nguyen
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Raymond Au
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Lachlan J Coin
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.,Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Infectious Disease, Imperial College London, London, London, UK
| | - Siok-Keen Tey
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia .,Department of Haematology and Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
21
|
Atkins A, Chung CH, Allen AG, Dampier W, Gurrola TE, Sariyer IK, Nonnemacher MR, Wigdahl B. Off-Target Analysis in Gene Editing and Applications for Clinical Translation of CRISPR/Cas9 in HIV-1 Therapy. Front Genome Ed 2021; 3:673022. [PMID: 34713260 PMCID: PMC8525399 DOI: 10.3389/fgeed.2021.673022] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022] Open
Abstract
As genome-editing nucleases move toward broader clinical applications, the need to define the limits of their specificity and efficiency increases. A variety of approaches for nuclease cleavage detection have been developed, allowing a full-genome survey of the targeting landscape and the detection of a variety of repair outcomes for nuclease-induced double-strand breaks. Each approach has advantages and disadvantages relating to the means of target-site capture, target enrichment mechanism, cellular environment, false discovery, and validation of bona fide off-target cleavage sites in cells. This review examines the strengths, limitations, and origins of the different classes of off-target cleavage detection systems including anchored primer enrichment (GUIDE-seq), in situ detection (BLISS), in vitro selection libraries (CIRCLE-seq), chromatin immunoprecipitation (ChIP) (DISCOVER-Seq), translocation sequencing (LAM PCR HTGTS), and in vitro genomic DNA digestion (Digenome-seq and SITE-Seq). Emphasis is placed on the specific modifications that give rise to the enhanced performance of contemporary techniques over their predecessors and the comparative performance of techniques for different applications. The clinical relevance of these techniques is discussed in the context of assessing the safety of novel CRISPR/Cas9 HIV-1 curative strategies. With the recent success of HIV-1 and SIV-1 viral suppression in humanized mice and non-human primates, respectively, using CRISPR/Cas9, rigorous exploration of potential off-target effects is of critical importance. Such analyses would benefit from the application of the techniques discussed in this review.
Collapse
Affiliation(s)
- Andrew Atkins
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States,Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Cheng-Han Chung
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States,Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Alexander G. Allen
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States,Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Will Dampier
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States,Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Theodore E. Gurrola
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States,Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ilker K. Sariyer
- Department of Neuroscience and Center for Neurovirology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Michael R. Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States,Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States,Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States,*Correspondence: Brian Wigdahl
| |
Collapse
|
22
|
Bode D, Cull AH, Rubio-Lara JA, Kent DG. Exploiting Single-Cell Tools in Gene and Cell Therapy. Front Immunol 2021; 12:702636. [PMID: 34322133 PMCID: PMC8312222 DOI: 10.3389/fimmu.2021.702636] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Single-cell molecular tools have been developed at an incredible pace over the last five years as sequencing costs continue to drop and numerous molecular assays have been coupled to sequencing readouts. This rapid period of technological development has facilitated the delineation of individual molecular characteristics including the genome, transcriptome, epigenome, and proteome of individual cells, leading to an unprecedented resolution of the molecular networks governing complex biological systems. The immense power of single-cell molecular screens has been particularly highlighted through work in systems where cellular heterogeneity is a key feature, such as stem cell biology, immunology, and tumor cell biology. Single-cell-omics technologies have already contributed to the identification of novel disease biomarkers, cellular subsets, therapeutic targets and diagnostics, many of which would have been undetectable by bulk sequencing approaches. More recently, efforts to integrate single-cell multi-omics with single cell functional output and/or physical location have been challenging but have led to substantial advances. Perhaps most excitingly, there are emerging opportunities to reach beyond the description of static cellular states with recent advances in modulation of cells through CRISPR technology, in particular with the development of base editors which greatly raises the prospect of cell and gene therapies. In this review, we provide a brief overview of emerging single-cell technologies and discuss current developments in integrating single-cell molecular screens and performing single-cell multi-omics for clinical applications. We also discuss how single-cell molecular assays can be usefully combined with functional data to unpick the mechanism of cellular decision-making. Finally, we reflect upon the introduction of spatial transcriptomics and proteomics, its complementary role with single-cell RNA sequencing (scRNA-seq) and potential application in cellular and gene therapy.
Collapse
Affiliation(s)
- Daniel Bode
- Wellcome Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Alyssa H. Cull
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Juan A. Rubio-Lara
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - David G. Kent
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| |
Collapse
|
23
|
Kok YL, Vongrad V, Chaudron SE, Shilaih M, Leemann C, Neumann K, Kusejko K, Di Giallonardo F, Kuster H, Braun DL, Kouyos RD, Günthard HF, Metzner KJ. HIV-1 integration sites in CD4+ T cells during primary, chronic, and late presentation of HIV-1 infection. JCI Insight 2021; 6:143940. [PMID: 33784259 PMCID: PMC8262285 DOI: 10.1172/jci.insight.143940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/25/2021] [Indexed: 12/29/2022] Open
Abstract
HIV-1 is capable of integrating its genome into that of its host cell. We examined the influence of the activation state of CD4+ T cells, the effect of antiretroviral therapy (ART), and the clinical stage of HIV-1 infection on HIV-1 integration site features and selection. HIV-1 integration sites were sequenced from longitudinally sampled resting and activated CD4+ T cells from 12 HIV-1–infected individuals. In total, 589 unique HIV-1 integration sites were analyzed: 147, 391, and 51 during primary, chronic, and late presentation of HIV-1 infection, respectively. As early as during primary HIV-1 infection and independent of the activation state of CD4+ T cells collected on and off ART, HIV-1 integration sites were preferentially detected in recurrent integration genes, genes associated with clonal expansion of latently HIV-1–infected CD4+ T cells, cancer-related genes, and highly expressed genes. The preference for cancer-related genes was more pronounced at late stages of HIV-1 infection. Host genomic features of HIV-1 integration site selection remained stable during HIV-1 infection in both resting and activated CD4+ T cells. In summary, characteristic HIV-1 integration site features are preestablished as early as during primary HIV-1 infection and are found in both resting and activated CD4+ T cells.
Collapse
Affiliation(s)
- Yik Lim Kok
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Valentina Vongrad
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Sandra E Chaudron
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Mohaned Shilaih
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Christine Leemann
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Kathrin Neumann
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Katharina Kusejko
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Francesca Di Giallonardo
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Herbert Kuster
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Dominique L Braun
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Roger D Kouyos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Karin J Metzner
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, and.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Bougnères P, Hacein-Bey-Abina S, Labik I, Adamsbaum C, Castaignède C, Bellesme C, Schmidt M. Long-Term Follow-Up of Hematopoietic Stem-Cell Gene Therapy for Cerebral Adrenoleukodystrophy. Hum Gene Ther 2021; 32:1260-1269. [PMID: 33789438 DOI: 10.1089/hum.2021.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In 2009, cerebral adrenoleukodystrophy (c-ALD) became the first brain disease to be treated with lentiviral (LV)-based hematopoietic stem cell gene therapy with the ABCD1 gene in four boys (P1-P4) who had demyelinating lesions expected to be lethal in the short term and no bone marrow donor. We report the clinical and magnetic resonance imaging (MRI) follow-up over a mean of 8.8 years posttransplant. In parallel, vector genome copies, expression of transgenic ALD protein (ALDP), and viral integration sites were determined in peripheral blood cells. Prior to transplant, the four patients had a normal or near normal neurocognitive status but gadolinium-enhanced demyelination in various brain regions. Gadolinium diffusion disappeared during the first year posttransplant. P3 kept a near normal status until 8.3 years of follow-up, but P1, P2, and P4 showed major cognitive degradation around 9, 28, and 60 months posttransplant. Neurological status and demyelination stabilized until last evaluation in P2, but deteriorated in both P1 at 10 years and P4 at 3 years posttransplant. The proportion of myeloid and lymphoid cells expressing transgenic ALDP decreased by half within 5 years then stabilized around 5% to 10%. Integration site analysis revealed a durable polyclonal distribution of genetically corrected hematopoietic cells. No adverse effects were observed. The long-term arrest of demyelination at MRI and persistence of transduced hematopoietic progenitors support that LV gene therapy may be a safe and durable treatment of c-ALD. However, the neurological degradation observed in three out of four patients mitigates the benefit of this therapy, calling for an earlier intervention, more potent vectors, and additional therapeutic strategies.
Collapse
Affiliation(s)
- Pierre Bougnères
- UMR1195 INSERM, Le Kremlin Bicêtre, France.,Université Paris Saclay, MIRCen Institute/Neuratris, Fontenay-aux-Roses, France.,Therapy Design Consulting, Vincennes, France
| | - Salima Hacein-Bey-Abina
- Clinical Immunology Laboratory, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Université Paris Saclay, Paris, France.,UTCBS, CNRS UMR8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Le Kremlin-Bicêtre, France
| | | | | | - Clémence Castaignède
- Pediatric Neurology, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Céline Bellesme
- Pediatric Neurology, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | | |
Collapse
|
25
|
van Haasteren J, Munis AM, Gill DR, Hyde SC. Genome-wide integration site detection using Cas9 enriched amplification-free long-range sequencing. Nucleic Acids Res 2021; 49:e16. [PMID: 33290561 PMCID: PMC7897500 DOI: 10.1093/nar/gkaa1152] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 10/13/2020] [Accepted: 11/10/2020] [Indexed: 12/31/2022] Open
Abstract
The gene and cell therapy fields are advancing rapidly, with a potential to treat and cure a wide range of diseases, and lentivirus-based gene transfer agents are the vector of choice for many investigators. Early cases of insertional mutagenesis caused by gammaretroviral vectors highlighted that integration site (IS) analysis was a major safety and quality control checkpoint for lentiviral applications. The methods established to detect lentiviral integrations using next-generation sequencing (NGS) are limited by short read length, inadvertent PCR bias, low yield, or lengthy protocols. Here, we describe a new method to sequence IS using Amplification-free Integration Site sequencing (AFIS-Seq). AFIS-Seq is based on amplification-free, Cas9-mediated enrichment of high-molecular-weight chromosomal DNA suitable for long-range Nanopore MinION sequencing. This accessible and low-cost approach generates long reads enabling IS mapping with high certainty within a single day. We demonstrate proof-of-concept by mapping IS of lentiviral vectors in a variety of cell models and report up to 1600-fold enrichment of the signal. This method can be further extended to sequencing of Cas9-mediated integration of genes and to in vivo analysis of IS. AFIS-Seq uses long-read sequencing to facilitate safety evaluation of preclinical lentiviral vector gene therapies by providing IS analysis with improved confidence.
Collapse
Affiliation(s)
- Joost van Haasteren
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Altar M Munis
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Deborah R Gill
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen C Hyde
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Xydia M, Rahbari R, Ruggiero E, Macaulay I, Tarabichi M, Lohmayer R, Wilkening S, Michels T, Brown D, Vanuytven S, Mastitskaya S, Laidlaw S, Grabe N, Pritsch M, Fronza R, Hexel K, Schmitt S, Müller-Steinhardt M, Halama N, Domschke C, Schmidt M, von Kalle C, Schütz F, Voet T, Beckhove P. Common clonal origin of conventional T cells and induced regulatory T cells in breast cancer patients. Nat Commun 2021; 12:1119. [PMID: 33602930 PMCID: PMC7893042 DOI: 10.1038/s41467-021-21297-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Regulatory CD4+ T cells (Treg) prevent tumor clearance by conventional T cells (Tconv) comprising a major obstacle of cancer immune-surveillance. Hitherto, the mechanisms of Treg repertoire formation in human cancers remain largely unclear. Here, we analyze Treg clonal origin in breast cancer patients using T-Cell Receptor and single-cell transcriptome sequencing. While Treg in peripheral blood and breast tumors are clonally distinct, Tconv clones, including tumor-antigen reactive effectors (Teff), are detected in both compartments. Tumor-infiltrating CD4+ cells accumulate into distinct transcriptome clusters, including early activated Tconv, uncommitted Teff, Th1 Teff, suppressive Treg and pro-tumorigenic Treg. Trajectory analysis suggests early activated Tconv differentiation either into Th1 Teff or into suppressive and pro-tumorigenic Treg. Importantly, Tconv, activated Tconv and Treg share highly-expanded clones contributing up to 65% of intratumoral Treg. Here we show that Treg in human breast cancer may considerably stem from antigen-experienced Tconv converting into secondary induced Treg through intratumoral activation.
Collapse
Affiliation(s)
- Maria Xydia
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany.
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany.
| | - Raheleh Rahbari
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
| | - Eliana Ruggiero
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Iain Macaulay
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- Technical Development, Earlham Institute, Norwich, UK
| | - Maxime Tarabichi
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- The Francis Crick Institute, London, UK
| | - Robert Lohmayer
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany
- Institute for Theoretical Physics, University of Regensburg, Regensburg, Germany
| | - Stefan Wilkening
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Tillmann Michels
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany
| | - Daniel Brown
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Sebastiaan Vanuytven
- The Francis Crick Institute, London, UK
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Svetlana Mastitskaya
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Sean Laidlaw
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
| | - Niels Grabe
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Hamamatsu Tissue Imaging and Analysis Centre, BIOQUANT, University of Heidelberg, Heidelberg, Germany
| | - Maria Pritsch
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany
| | - Raffaele Fronza
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Klaus Hexel
- Flow Cytometry Core Facility, German Cancer Research Centre, Heidelberg, Germany
| | - Steffen Schmitt
- Flow Cytometry Core Facility, German Cancer Research Centre, Heidelberg, Germany
| | - Michael Müller-Steinhardt
- German Red Cross (DRK Blood Donation Service in Baden-Württemberg-Hessen) and Institute for Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Niels Halama
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Hamamatsu Tissue Imaging and Analysis Centre, BIOQUANT, University of Heidelberg, Heidelberg, Germany
| | - Christoph Domschke
- Department of Gynecology and Obstetrics, University Hospital of Heidelberg, Heidelberg, Germany
| | - Manfred Schmidt
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Christof von Kalle
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
- Clinical Study Centre, Charité/BIH, Berlin, Germany
| | - Florian Schütz
- Department of Gynecology and Obstetrics, University Hospital of Heidelberg, Heidelberg, Germany
| | - Thierry Voet
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Philipp Beckhove
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany.
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany.
| |
Collapse
|
27
|
Asif M, Siddiqui HA, Naqvi RZ, Amin I, Asad S, Mukhtar Z, Bashir A, Mansoor S. Development of event-specific detection method for identification of insect resistant NIBGE-1601 cotton harboring double gene Cry1Ac-Cry2Ab construct. Sci Rep 2021; 11:3479. [PMID: 33568702 PMCID: PMC7876094 DOI: 10.1038/s41598-021-82798-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 01/07/2021] [Indexed: 11/09/2022] Open
Abstract
Bt cotton expressing Cry1Ac is being cultivated in Pakistan. It has been observed that pink bollworm may have developed resistance against single Bt gene (Cry1Ac). For durable resistance, insect resistant NIBGE-1601 cotton harboring double gene Cry1Ac-Cry2Ab construct was developed. There was a need to characterize NIBGE-1601 event for intellectual property rights protection. The Presence of NIBGE Cry1Ac and NIBGE Cry2Ab genes was checked in NIBGE-1601 cotton plants through PCR, while there was no amplification using primers specific for Monsanto events (MON531, MON15985, MON1445). Using genome walking technology, NIBGE-601 event has been characterized. Event-specific primers of NIBGE-1601 were designed and evaluated to differentiate it from other cotton events mentioned above. NIBGE-1601 event detection primers are highly specific, therefore, can detect NIBGE 1601 event at different conditions using single or multiplex PCR. In the qualitative PCR, using NIBGE-1601 event specific primers, 0.05 ng was the limit of detection for NIBGE-1601double gene cotton genomic DNA. Thus event characterization and development of event-specific diagnostics will help in breeding new cotton varieties resistant to cotton bollworms.
Collapse
Affiliation(s)
- Muhammad Asif
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic, Engineering, Faisalabad, Pakistan
| | - Hamid Anees Siddiqui
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic, Engineering, Faisalabad, Pakistan.,Pakistan Institute of Engineering and Applied Sciences, Nilore, Pakistan
| | - Rubab Zahra Naqvi
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic, Engineering, Faisalabad, Pakistan
| | - Imran Amin
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic, Engineering, Faisalabad, Pakistan
| | - Shaheen Asad
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic, Engineering, Faisalabad, Pakistan
| | - Zahid Mukhtar
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic, Engineering, Faisalabad, Pakistan
| | - Aftab Bashir
- Department of Biological Sciences, Forman Christian College, Lahore, Pakistan
| | - Shahid Mansoor
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic, Engineering, Faisalabad, Pakistan.
| |
Collapse
|
28
|
Wang X, Wu Y, Yee JK. Detection of CRISPR/Cas9-Generated Off-Target Effect by Integration-Defective Lentiviral Vector. Methods Mol Biol 2021; 2162:243-260. [PMID: 32926387 DOI: 10.1007/978-1-0716-0687-2_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR) and other gene editing technologies such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) show great promises for research and therapeutic applications. One major concern is the off-target effects generated by these nucleases at unintended genomic sequences. In silico methods are usually used for off-target site prediction. However, based on currently available algorithms, the predicted cleavage activity at these potential off-target sites does not always reflect the true cleavage in vivo. Here we present an unbiased screening protocol using integration-defective lentiviral vector (IDLV) and deep sequencing to map the off-target sites generated by gene editing tools.
Collapse
Affiliation(s)
- Xiaoling Wang
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Youjun Wu
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Jiing-Kuan Yee
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
29
|
Rosewick N, Hahaut V, Durkin K, Artesi M, Karpe S, Wayet J, Griebel P, Arsic N, Marçais A, Hermine O, Burny A, Georges M, Van den Broeke A. An Improved Sequencing-Based Bioinformatics Pipeline to Track the Distribution and Clonal Architecture of Proviral Integration Sites. Front Microbiol 2020; 11:587306. [PMID: 33193242 PMCID: PMC7606357 DOI: 10.3389/fmicb.2020.587306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022] Open
Abstract
The combined application of linear amplification-mediated PCR (LAM-PCR) protocols with next-generation sequencing (NGS) has had a large impact on our understanding of retroviral pathogenesis. Previously, considerable effort has been expended to optimize NGS methods to explore the genome-wide distribution of proviral integration sites and the clonal architecture of clinically important retroviruses like human T-cell leukemia virus type-1 (HTLV-1). Once sequencing data are generated, the application of rigorous bioinformatics analysis is central to the biological interpretation of the data. To better exploit the potential information available through these methods, we developed an optimized bioinformatics pipeline to analyze NGS clonality datasets. We found that short-read aligners, specifically designed to manage NGS datasets, provide increased speed, significantly reducing processing time and decreasing the computational burden. This is achieved while also accounting for sequencing base quality. We demonstrate the utility of an additional trimming step in the workflow, which adjusts for the number of reads supporting each insertion site. In addition, we developed a recall procedure to reduce bias associated with proviral integration within low complexity regions of the genome, providing a more accurate estimation of clone abundance. Finally, we recommend the application of a “clean-and-recover” step to clonality datasets generated from large cohorts and longitudinal studies. In summary, we report an optimized bioinformatics workflow for NGS clonality analysis and describe a new set of steps to guide the computational process. We demonstrate that the application of this protocol to the analysis of HTLV-1 and bovine leukemia virus (BLV) clonality datasets improves the quality of data processing and provides a more accurate definition of the clonal landscape in infected individuals. The optimized workflow and analysis recommendations can be implemented in the majority of bioinformatics pipelines developed to analyze LAM-PCR-based NGS clonality datasets.
Collapse
Affiliation(s)
- Nicolas Rosewick
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Unit of Animal Genomics, GIGA, Université de Liège (ULiège), Liège, Belgium
| | - Vincent Hahaut
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Unit of Animal Genomics, GIGA, Université de Liège (ULiège), Liège, Belgium
| | - Keith Durkin
- Unit of Animal Genomics, GIGA, Université de Liège (ULiège), Liège, Belgium
| | - Maria Artesi
- Unit of Animal Genomics, GIGA, Université de Liège (ULiège), Liège, Belgium
| | - Snehal Karpe
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Unit of Animal Genomics, GIGA, Université de Liège (ULiège), Liège, Belgium
| | - Jérôme Wayet
- Unit of Animal Genomics, GIGA, Université de Liège (ULiège), Liège, Belgium
| | - Philip Griebel
- VIDO-Intervac, University of Saskatchewan, Saskatoon, SK, Canada
| | - Natasa Arsic
- VIDO-Intervac, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ambroise Marçais
- Service d'hématologie, Hôpital Universitaire Necker, Université René Descartes, Assistance publique hôpitaux de Paris, Paris, France
| | - Olivier Hermine
- Service d'hématologie, Hôpital Universitaire Necker, Université René Descartes, Assistance publique hôpitaux de Paris, Paris, France.,Institut Imagine, INSERM U1163, CNRS ERL8654, Paris, France
| | - Arsène Burny
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Michel Georges
- Unit of Animal Genomics, GIGA, Université de Liège (ULiège), Liège, Belgium
| | - Anne Van den Broeke
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Unit of Animal Genomics, GIGA, Université de Liège (ULiège), Liège, Belgium
| |
Collapse
|
30
|
Gadgil RY, Romer EJ, Goodman CC, Rider SD, Damewood FJ, Barthelemy JR, Shin-Ya K, Hanenberg H, Leffak M. Replication stress at microsatellites causes DNA double-strand breaks and break-induced replication. J Biol Chem 2020; 295:15378-15397. [PMID: 32873711 DOI: 10.1074/jbc.ra120.013495] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 08/23/2020] [Indexed: 12/12/2022] Open
Abstract
Short tandemly repeated DNA sequences, termed microsatellites, are abundant in the human genome. These microsatellites exhibit length instability and susceptibility to DNA double-strand breaks (DSBs) due to their tendency to form stable non-B DNA structures. Replication-dependent microsatellite DSBs are linked to genome instability signatures in human developmental diseases and cancers. To probe the causes and consequences of microsatellite DSBs, we designed a dual-fluorescence reporter system to detect DSBs at expanded (CTG/CAG) n and polypurine/polypyrimidine (Pu/Py) mirror repeat structures alongside the c-myc replication origin integrated at a single ectopic chromosomal site. Restriction cleavage near the (CTG/CAG)100 microsatellite leads to homology-directed single-strand annealing between flanking AluY elements and reporter gene deletion that can be detected by flow cytometry. However, in the absence of restriction cleavage, endogenous and exogenous replication stressors induce DSBs at the (CTG/CAG)100 and Pu/Py microsatellites. DSBs map to a narrow region at the downstream edge of the (CTG)100 lagging-strand template. (CTG/CAG) n chromosome fragility is repeat length-dependent, whereas instability at the (Pu/Py) microsatellites depends on replication polarity. Strikingly, restriction-generated DSBs and replication-dependent DSBs are not repaired by the same mechanism. Knockdown of DNA damage response proteins increases (Rad18, polymerase (Pol) η, Pol κ) or decreases (Mus81) the sensitivity of the (CTG/CAG)100 microsatellites to replication stress. Replication stress and DSBs at the ectopic (CTG/CAG)100 microsatellite lead to break-induced replication and high-frequency mutagenesis at a flanking thymidine kinase gene. Our results show that non-B structure-prone microsatellites are susceptible to replication-dependent DSBs that cause genome instability.
Collapse
Affiliation(s)
- Rujuta Yashodhan Gadgil
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Eric J Romer
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Caitlin C Goodman
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - S Dean Rider
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - French J Damewood
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Joanna R Barthelemy
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Kazuo Shin-Ya
- Biomedical Information Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Helmut Hanenberg
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany; Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Leffak
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA.
| |
Collapse
|
31
|
Saito M, Hasegawa H, Yamauchi S, Nakagawa S, Sasaki D, Nao N, Tanio M, Wada Y, Matsudaira T, Momose H, Kuramitsu M, Yamagishi M, Nakashima M, Nakahata S, Iha H, Ogata M, Imaizumi Y, Uchimaru K, Morishita K, Watanabe T, Miyazaki Y, Yanagihara K. A high-throughput detection method for the clonality of Human T-cell leukemia virus type-1-infected cells in vivo. Int J Hematol 2020; 112:300-306. [PMID: 32725607 DOI: 10.1007/s12185-020-02935-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022]
Abstract
Approximately 10-20 million of Human T-cell leukemia virus type-1 (HTLV-1)-infected carriers have been previously reported, and approximately 5% of these carriers develop adult T-cell leukemia/lymphoma (ATL) with a characteristic poor prognosis. In Japan, Southern blotting has long been routinely performed for detection of clonally expanded ATL cells in vivo, and as a confirmatory diagnostic test for ATL. However, alternative methods to Southern blotting, such as sensitive, quantitative, and rapid analytical methods, are currently required in clinical practice. In this study, we developed a high-throughput method called rapid amplification of integration site (RAIS) that could amplify HTLV-1-integrated fragments within 4 h and detect the integration sites in > 0.16% of infected cells. Furthermore, we established a novel quantification method for HTLV-1 clonality using Sanger sequencing with RAIS products, and the validity of the quantification method was confirmed by comparing it with next-generation sequencing in terms of the clonality. Thus, we believe that RAIS has a high potential for use as an alternative routine molecular confirmatory test for the clonality analysis of HTLV-1-infected cells.
Collapse
Affiliation(s)
- Masumichi Saito
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan.
| | - Hiroo Hasegawa
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, Japan. .,Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Shunsuke Yamauchi
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - So Nakagawa
- Biomedical Informatics Laboratory, Department of Molecular Life Science, Tokai University School of Medicine, Kanagawa, Japan.,Micro/Nano Technology Center, Tokai University, Kanagawa, Japan
| | - Daisuke Sasaki
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Naganori Nao
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Michikazu Tanio
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yusaku Wada
- Biotechnological Research Support Division, FASMAC Co., Ltd, Kanagawa, Japan
| | - Takahiro Matsudaira
- Biotechnological Research Support Division, FASMAC Co., Ltd, Kanagawa, Japan
| | - Haruka Momose
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Madoka Kuramitsu
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Yamagishi
- Graduate School of Frontier Sciences, Department of Computational Biology and Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Makoto Nakashima
- Graduate School of Frontier Sciences, Department of Computational Biology and Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shingo Nakahata
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, Miyazaki, Japan
| | - Hidekatsu Iha
- Department of Microbiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masao Ogata
- Department of Hematology, Oita University, Oita, Japan
| | | | - Kaoru Uchimaru
- Graduate School of Frontier Sciences, Department of Computational Biology and Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Morishita
- Division of Tumor and Cellular Biochemistry, Department of Medical Sciences, University of Miyazaki, Miyazaki, Japan
| | - Toshiki Watanabe
- The Institute of Medical Science Research Hospital and Future Center Initiative, The University of Tokyo, Tokyo, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan.,Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, Japan.,Department of Laboratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
32
|
McHugh D, Myburgh R, Caduff N, Spohn M, Kok YL, Keller CW, Murer A, Chatterjee B, Rühl J, Engelmann C, Chijioke O, Quast I, Shilaih M, Strouvelle VP, Neumann K, Menter T, Dirnhofer S, Lam JK, Hui KF, Bredl S, Schlaepfer E, Sorce S, Zbinden A, Capaul R, Lünemann JD, Aguzzi A, Chiang AK, Kempf W, Trkola A, Metzner KJ, Manz MG, Grundhoff A, Speck RF, Münz C. EBV renders B cells susceptible to HIV-1 in humanized mice. Life Sci Alliance 2020; 3:3/8/e202000640. [PMID: 32576602 PMCID: PMC7335381 DOI: 10.26508/lsa.202000640] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
HIV and EBV are human pathogens that cause a considerable burden to worldwide health. In combination, these viruses are linked to AIDS-associated lymphomas. We found that EBV, which transforms B cells, renders them susceptible to HIV-1 infection in a CXCR4 and CD4-dependent manner in vitro and that CXCR4-tropic HIV-1 integrates into the genome of these B cells with the same molecular profile as in autologous CD4+ T cells. In addition, we established a humanized mouse model to investigate the in vivo interactions of EBV and HIV-1 upon coinfection. The respective mice that reconstitute human immune system components upon transplantation with CD34+ human hematopoietic progenitor cells could recapitulate aspects of EBV and HIV immunobiology observed in dual-infected patients. Upon coinfection of humanized mice, EBV/HIV dual-infected B cells could be detected, but were susceptible to CD8+ T-cell-mediated immune control.
Collapse
Affiliation(s)
- Donal McHugh
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Renier Myburgh
- Department of Medical Oncology and Hematology, University and University Hospital of Zürich, Zürich, Switzerland
| | - Nicole Caduff
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Michael Spohn
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Yik Lim Kok
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland.,Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Christian W Keller
- Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Anita Murer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Bithi Chatterjee
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Julia Rühl
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Christine Engelmann
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Obinna Chijioke
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.,Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| | - Isaak Quast
- Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Mohaned Shilaih
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Victoria P Strouvelle
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland.,Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Kathrin Neumann
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Thomas Menter
- Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| | - Stephan Dirnhofer
- Institute of Pathology and Medical Genetics, University Hospital of Basel, Basel, Switzerland
| | - Janice Kp Lam
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kwai F Hui
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Simon Bredl
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Erika Schlaepfer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Silvia Sorce
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Andrea Zbinden
- Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Riccarda Capaul
- Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Jan D Lünemann
- Neuroinflammation, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, Zurich, Switzerland
| | - Alan Ks Chiang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Werner Kempf
- Kempf und Pfaltz Histologische Diagnostik AG, Zürich, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Karin J Metzner
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland.,Institute of Medical Virology, University of Zürich, Zürich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University and University Hospital of Zürich, Zürich, Switzerland
| | - Adam Grundhoff
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Roberto F Speck
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Zürich, Zürich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
33
|
Breton C, Clark PM, Wang L, Greig JA, Wilson JM. ITR-Seq, a next-generation sequencing assay, identifies genome-wide DNA editing sites in vivo following adeno-associated viral vector-mediated genome editing. BMC Genomics 2020; 21:239. [PMID: 32183699 PMCID: PMC7076944 DOI: 10.1186/s12864-020-6655-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Identifying nuclease-induced double-stranded breaks in DNA on a genome-wide scale is critical for assessing the safety and efficacy of genome editing therapies. We previously demonstrated that after administering adeno-associated viral (AAV) vector-mediated genome-editing strategies in vivo, vector sequences integrated into the host organism's genomic DNA at double-stranded breaks. Thus, identifying the genomic location of inserted AAV sequences would enable us to identify DSB events, mainly derived from the nuclease on- and off-target activity. RESULTS Here, we developed a next-generation sequencing assay that detects insertions of specific AAV vector sequences called inverted terminal repeats (ITRs). This assay, ITR-Seq, enables us to identify off-target nuclease activity in vivo. Using ITR-Seq, we analyzed liver DNA samples of rhesus macaques treated with AAV vectors expressing a meganuclease. We found dose-dependent off-target activity and reductions in off-target events induced by further meganuclease development. In mice, we identified the genomic locations of ITR integration after treatment with Cas9 nucleases and their corresponding single-guide RNAs. CONCLUSIONS In sum, ITR-Seq is a powerful method for identifying off-target sequences induced by AAV vector-delivered genome-editing nucleases. ITR-Seq will help us understand the specificity and efficacy of different genome-editing nucleases in animal models and clinical studies. This information can help enhance the safety profile of gene-editing therapies.
Collapse
Affiliation(s)
- Camilo Breton
- Gene Therapy Program, University of Pennsylvania Perelman School of Medicine, 125 South 31st Street, Suite 1200, Philadelphia, PA, 19104, USA
| | - Peter M Clark
- Gene Therapy Program, University of Pennsylvania Perelman School of Medicine, 125 South 31st Street, Suite 1200, Philadelphia, PA, 19104, USA
| | - Lili Wang
- Gene Therapy Program, University of Pennsylvania Perelman School of Medicine, 125 South 31st Street, Suite 1200, Philadelphia, PA, 19104, USA
| | - Jenny A Greig
- Gene Therapy Program, University of Pennsylvania Perelman School of Medicine, 125 South 31st Street, Suite 1200, Philadelphia, PA, 19104, USA
| | - James M Wilson
- Gene Therapy Program, University of Pennsylvania Perelman School of Medicine, 125 South 31st Street, Suite 1200, Philadelphia, PA, 19104, USA.
| |
Collapse
|
34
|
Straetemans T, Janssen A, Jansen K, Doorn R, Aarts T, van Muyden ADD, Simonis M, Bergboer J, de Witte M, Sebestyen Z, Kuball J. TEG001 Insert Integrity from Vector Producer Cells until Medicinal Product. Mol Ther 2019; 28:561-571. [PMID: 31882320 DOI: 10.1016/j.ymthe.2019.11.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 12/20/2022] Open
Abstract
Despite extensive usage of gene therapy medicinal products (GTMPs) in clinical studies and recent approval of chimeric antigen receptor (CAR) T cell therapy, little information has been made available on the precise molecular characterization and possible variations in terms of insert integrity and vector copy numbers of different GTMPs during the complete production chain. Within this context, we characterize αβT cells engineered to express a defined γδT cell engineered to express a defined γδT receptor (TEG) currently used in a first-in-human clinical study (NTR6541). Utilizing targeted locus amplification in combination with next generation sequencing for the vector producer clone and TEG001 products, we report on five single-nucleotide variants and nine intact vector copies integrated in the producer clone. The vector copy number in TEG001 cells was on average a factor 0.72 (SD 0.11) below that of the producer cell clone. All nucleotide variants were transferred to TEG001 without having an effect on cellular proliferation during extensive in vitro culture. Based on an environmental risk assessment of the five nucleotide variants present in the non-coding viral region of the TEG001 insert, there was no altered environmental impact of TEG001 cells. We conclude that TEG001 cells do not have an increased risk for malignant transformation in vivo.
Collapse
Affiliation(s)
- Trudy Straetemans
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Anke Janssen
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Koen Jansen
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ruud Doorn
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Tineke Aarts
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Anna D D van Muyden
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | | | - Moniek de Witte
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Zsolt Sebestyen
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jurgen Kuball
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
35
|
Handumrongkul C, Ye AL, Chmura SA, Soroceanu L, Mack M, Ice RJ, Thistle R, Myers M, Ursu SJ, Liu Y, Kashani-Sabet M, Heath TD, Liggitt D, Lewis DB, Debs R. Durable multitransgene expression in vivo using systemic, nonviral DNA delivery. SCIENCE ADVANCES 2019; 5:eaax0217. [PMID: 31807699 PMCID: PMC6881169 DOI: 10.1126/sciadv.aax0217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 10/02/2019] [Indexed: 05/05/2023]
Abstract
Recombinant adeno-associated virus (AAV) vectors are transforming therapies for rare human monogenic deficiency diseases. However, adaptive immune responses to AAV and its limited DNA insert capacity, restrict their therapeutic potential. HEDGES (high-level extended duration gene expression system), a nonviral DNA- and liposome-based gene delivery platform, overcomes these limitations in immunocompetent mice. Specifically, one systemic HEDGES injection durably produces therapeutic levels of transgene-encoded human proteins, including FDA-approved cytokines and monoclonal antibodies, without detectable integration into genomic DNA. HEDGES also controls protein production duration from <3 weeks to >1.5 years, does not induce anti-vector immune responses, is reexpressed for prolonged periods following reinjection, and produces only transient minimal toxicity. HEDGES can produce extended therapeutic levels of multiple transgene-encoded therapeutic human proteins from DNA inserts >1.5-fold larger than AAV-based therapeutics, thus creating combinatorial interventions to effectively treat common polygenic diseases driven by multigenic abnormalities.
Collapse
Affiliation(s)
| | | | | | - Liliana Soroceanu
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | | | - Ryan J. Ice
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Robert Thistle
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | | | - Sarah J. Ursu
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Yong Liu
- DNARx LLC, San Francisco, CA, USA
| | | | | | - Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - David B. Lewis
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Robert Debs
- DNARx LLC, San Francisco, CA, USA
- Corresponding author.
| |
Collapse
|
36
|
Josi C, Bürki S, Vidal S, Dordet-Frisoni E, Citti C, Falquet L, Pilo P. Large-Scale Analysis of the Mycoplasma bovis Genome Identified Non-essential, Adhesion- and Virulence-Related Genes. Front Microbiol 2019; 10:2085. [PMID: 31572317 PMCID: PMC6753880 DOI: 10.3389/fmicb.2019.02085] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/23/2019] [Indexed: 12/21/2022] Open
Abstract
Mycoplasma bovis is an important pathogen of cattle causing bovine mycoplasmosis. Clinical manifestations are numerous, but pneumonia, mastitis, and arthritis cases are mainly reported. Currently, no efficient vaccine is available and antibiotic treatments are not always satisfactory. The design of new, efficient prophylactic and therapeutic approaches requires a better understanding of the molecular mechanisms responsible for M. bovis pathogenicity. Random transposon mutagenesis has been widely used in Mycoplasma species to identify potential gene functions. Such an approach can also be used to screen genomes and search for essential and non-essential genes for growth. Here, we generated a random transposon mutant library of M. bovis strain JF4278 containing approximately 4000 independent insertion sites. We then coupled high-throughput screening of this mutant library to transposon sequencing and bioinformatic analysis to identify M. bovis non-essential, adhesion- and virulence-related genes. Three hundred and fifty-two genes of M. bovis were assigned as essential for growth in rich medium. Among the remaining non-essential genes, putative virulence-related factors were subsequently identified. The complete mutant library was screened for adhesion using primary bovine mammary gland epithelial cells. Data from this assay resulted in a list of conditional-essential genes with putative adhesion-related functions by identifying non-essential genes for growth that are essential for host cell-adhesion. By individually assessing the adhesion capacity of six selected mutants, two previously unknown factors and the adhesin TrmFO were associated with a reduced adhesion phenotype. Overall, our study (i) uncovers new, putative virulence-related genes; (ii) offers a list of putative adhesion-related factors; and (iii) provides valuable information for vaccine design and for exploring M. bovis biology, pathogenesis, and host-interaction.
Collapse
Affiliation(s)
- Christoph Josi
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sibylle Bürki
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland
| | - Sara Vidal
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland
| | | | - Christine Citti
- UMR 1225, IHAP, Université de Toulouse, INRA, ENVT, Toulouse, France
| | - Laurent Falquet
- Department of Biology, Faculty of Science and Medicine, Swiss Institute of Bioinformatics, University of Fribourg, Fribourg, Switzerland
| | - Paola Pilo
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland
| |
Collapse
|
37
|
Río P, Navarro S, Wang W, Sánchez-Domínguez R, Pujol RM, Segovia JC, Bogliolo M, Merino E, Wu N, Salgado R, Lamana ML, Yañez RM, Casado JA, Giménez Y, Román-Rodríguez FJ, Álvarez L, Alberquilla O, Raimbault A, Guenechea G, Lozano ML, Cerrato L, Hernando M, Gálvez E, Hladun R, Giralt I, Barquinero J, Galy A, García de Andoín N, López R, Catalá A, Schwartz JD, Surrallés J, Soulier J, Schmidt M, Díaz de Heredia C, Sevilla J, Bueren JA. Successful engraftment of gene-corrected hematopoietic stem cells in non-conditioned patients with Fanconi anemia. Nat Med 2019; 25:1396-1401. [PMID: 31501599 DOI: 10.1038/s41591-019-0550-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/18/2019] [Indexed: 12/20/2022]
Abstract
Fanconi anemia (FA) is a DNA repair syndrome generated by mutations in any of the 22 FA genes discovered to date1,2. Mutations in FANCA account for more than 60% of FA cases worldwide3,4. Clinically, FA is associated with congenital abnormalities and cancer predisposition. However, bone marrow failure is the primary pathological feature of FA that becomes evident in 70-80% of patients with FA during the first decade of life5,6. In this clinical study (ClinicalTrials.gov, NCT03157804 ; European Clinical Trials Database, 2011-006100-12), we demonstrate that lentiviral-mediated hematopoietic gene therapy reproducibly confers engraftment and proliferation advantages of gene-corrected hematopoietic stem cells (HSCs) in non-conditioned patients with FA subtype A. Insertion-site analyses revealed the multipotent nature of corrected HSCs and showed that the repopulation advantage of these cells was not due to genotoxic integrations of the therapeutic provirus. Phenotypic correction of blood and bone marrow cells was shown by the acquired resistance of hematopoietic progenitors and T lymphocytes to DNA cross-linking agents. Additionally, an arrest of bone marrow failure progression was observed in patients with the highest levels of gene marking. The progressive engraftment of corrected HSCs in non-conditioned patients with FA supports that gene therapy should constitute an innovative low-toxicity therapeutic option for this life-threatening disorder.
Collapse
Affiliation(s)
- Paula Río
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Susana Navarro
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Wei Wang
- Division of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany.,GeneWerk, Heidelberg, Germany
| | - Rebeca Sánchez-Domínguez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Roser M Pujol
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Instituto de Investigaciones Biomédicas, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - José C Segovia
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Massimo Bogliolo
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Instituto de Investigaciones Biomédicas, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Eva Merino
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Servicio de Hematología y Oncología Pediátrica, Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Ning Wu
- Division of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Rocío Salgado
- Servicio de Hematología, Hospital Universitario Fundación Jiménez Diaz, Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - María L Lamana
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Rosa M Yañez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - José A Casado
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Yari Giménez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Francisco J Román-Rodríguez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Lara Álvarez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Omaira Alberquilla
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Anna Raimbault
- Université de Paris (IRSL, INSERM, CNRS), Paris, France.,Hôpital Saint-Louis, Paris, France
| | - Guillermo Guenechea
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - M Luz Lozano
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Laura Cerrato
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Miriam Hernando
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Eva Gálvez
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Servicio de Hematología y Oncología Pediátrica, Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Raquel Hladun
- Servicio de Oncología y Hematología Pediátricas, Hospital Universitari Vall d'Hebron, Barcelona, Spain.,Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Irina Giralt
- Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | | | | | | | - Ricardo López
- Osakidetza Basque Health Service, Pediatric Oncology and Hematology Unit, Cruces University Hospital, Barakaldo, Spain
| | - Albert Catalá
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Servicio de Hematología y Oncología, Hospital Sant Joan de Déu, Barcelona, Spain
| | | | - Jordi Surrallés
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Instituto de Investigaciones Biomédicas, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jean Soulier
- Université de Paris (IRSL, INSERM, CNRS), Paris, France.,Hôpital Saint-Louis, Paris, France
| | - Manfred Schmidt
- Division of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany.,GeneWerk, Heidelberg, Germany
| | - Cristina Díaz de Heredia
- Servicio de Oncología y Hematología Pediátricas, Hospital Universitari Vall d'Hebron, Barcelona, Spain.,Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Julián Sevilla
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Servicio de Hematología y Oncología Pediátrica, Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Juan A Bueren
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain. .,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain.
| |
Collapse
|
38
|
Di WL, Lwin SM, Petrova A, Bernadis C, Syed F, Farzaneh F, Moulding D, Martinez AE, Sebire NJ, Rampling D, Virasami A, Zamiri M, Wang W, Hara H, Kadiyirire T, Abdul-Wahab A, Martinez-Queipo M, Harper JI, McGrath JA, Thrasher AJ, Mellerio JE, Qasim W. Generation and Clinical Application of Gene-Modified Autologous Epidermal Sheets in Netherton Syndrome: Lessons Learned from a Phase 1 Trial. Hum Gene Ther 2019; 30:1067-1078. [PMID: 31288584 DOI: 10.1089/hum.2019.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Netherton syndrome (NS) is a rare autosomal recessive skin disorder caused by mutations in SPINK5. It is a debilitating condition with notable mortality in the early years of life. There is no curative treatment. We undertook a nonrandomized, open-label, feasibility, and safety study using autologous keratinocytes transduced with a lentiviral vector encoding SPINK5 under the control of the human involucrin promoter. Six NS subjects were recruited, and gene-modified epithelial sheets were successfully generated in three of five subjects. The sheets exhibited expression of correctly sized lympho-epithelial Kazal-type-related inhibitor (LEKTI) protein after modification. One subject was grafted with a 20 cm2 gene-modified graft on the left anterior thigh without any adverse complications and was monitored by serial sampling for 12 months. Recovery within the graft area was compared against an area outside by morphology, proviral copy number and expression of the SPINK5 encoded protein, LEKTI, and its downstream target kallikrein 5, which exhibited transient functional correction. The study confirmed the feasibility of generating lentiviral gene-modified epidermal sheets for inherited skin diseases such as NS, but sustained LEKTI expression is likely to require the identification, targeting, and engraftment of long-lived keratinocyte stem cell populations for durable therapeutic effects. Important learning points for the application of gene-modified epidermal sheets are discussed.
Collapse
Affiliation(s)
- Wei-Li Di
- Infection, Immunity and Inflammation Programme, UCL GOS Institute of Child Health, London, United Kingdom
| | - Su M Lwin
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, United Kingdom
| | - Anastasia Petrova
- Infection, Immunity and Inflammation Programme, UCL GOS Institute of Child Health, London, United Kingdom
| | - Catina Bernadis
- Plastic Surgery Unit, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Farhatullah Syed
- Infection, Immunity and Inflammation Programme, UCL GOS Institute of Child Health, London, United Kingdom
| | - Farzin Farzaneh
- Cell and Gene Therapy-King's (CGT-K), School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Dale Moulding
- Light Microscopy Core Facility, UCL GOS Institute of Child Health, London, United Kingdom
| | - Anna E Martinez
- Dermatology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Neil J Sebire
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Dyanne Rampling
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Alex Virasami
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Mozheh Zamiri
- School of Life Sciences, University of Dundee, Scotland, United Kingdom
| | - Wei Wang
- Department of Translational Oncology, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Heidelberg, Germany.,GeneWerk GmbH, Heidelberg, Germany
| | - Havinder Hara
- Infection, Immunity and Inflammation Programme, UCL GOS Institute of Child Health, London, United Kingdom
| | - Tendai Kadiyirire
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, United Kingdom
| | - Alya Abdul-Wahab
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, United Kingdom
| | | | - John I Harper
- Infection, Immunity and Inflammation Programme, UCL GOS Institute of Child Health, London, United Kingdom
| | - John A McGrath
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, United Kingdom
| | - Adrian J Thrasher
- Infection, Immunity and Inflammation Programme, UCL GOS Institute of Child Health, London, United Kingdom
| | - Jemima E Mellerio
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, United Kingdom
| | - Waseem Qasim
- Infection, Immunity and Inflammation Programme, UCL GOS Institute of Child Health, London, United Kingdom
| |
Collapse
|
39
|
Slesarev A, Viswanathan L, Tang Y, Borgschulte T, Achtien K, Razafsky D, Onions D, Chang A, Cote C. CRISPR/CAS9 targeted CAPTURE of mammalian genomic regions for characterization by NGS. Sci Rep 2019; 9:3587. [PMID: 30837529 PMCID: PMC6401131 DOI: 10.1038/s41598-019-39667-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 01/30/2019] [Indexed: 01/08/2023] Open
Abstract
The robust detection of structural variants in mammalian genomes remains a challenge. It is particularly difficult in the case of genetically unstable Chinese hamster ovary (CHO) cell lines with only draft genome assemblies available. We explore the potential of the CRISPR/Cas9 system for the targeted capture of genomic loci containing integrated vectors in CHO-K1-based cell lines followed by next generation sequencing (NGS), and compare it to popular target-enrichment sequencing methods and to whole genome sequencing (WGS). Three different CRISPR/Cas9-based techniques were evaluated; all of them allow for amplification-free enrichment of target genomic regions in the range from 5 to 60 fold, and for recovery of ~15 kb-long sequences with no sequencing artifacts introduced. The utility of these protocols has been proven by the identification of transgene integration sites and flanking sequences in three CHO cell lines. The long enriched fragments helped to identify Escherichia coli genome sequences co-integrated with vectors, and were further characterized by Whole Genome Sequencing (WGS). Other advantages of CRISPR/Cas9-based methods are the ease of bioinformatics analysis, potential for multiplexing, and the production of long target templates for real-time sequencing.
Collapse
Affiliation(s)
- Alexei Slesarev
- BioReliance Corp., 14920 Broschart Road, Rockville, MD, 20850, USA.
| | | | - Yitao Tang
- BioReliance Corp., 14920 Broschart Road, Rockville, MD, 20850, USA
| | | | | | - David Razafsky
- MilliporeSigma, 2909 Laclede Avenue, Saint Louis, MO, 63103, USA
| | - David Onions
- BioReliance Corp., 14920 Broschart Road, Rockville, MD, 20850, USA
| | - Audrey Chang
- BioReliance Corp., 14920 Broschart Road, Rockville, MD, 20850, USA
| | - Colette Cote
- BioReliance Corp., 14920 Broschart Road, Rockville, MD, 20850, USA
| |
Collapse
|
40
|
Einkauf KB, Lee GQ, Gao C, Sharaf R, Sun X, Hua S, Chen SM, Jiang C, Lian X, Chowdhury FZ, Rosenberg ES, Chun TW, Li JZ, Yu XG, Lichterfeld M. Intact HIV-1 proviruses accumulate at distinct chromosomal positions during prolonged antiretroviral therapy. J Clin Invest 2019; 129:988-998. [PMID: 30688658 DOI: 10.1172/jci124291] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/04/2018] [Indexed: 12/16/2022] Open
Abstract
Chromosomal integration of genome-intact HIV-1 sequences into the host genome creates a reservoir of virally infected cells that persists throughout life, necessitating indefinite antiretroviral suppression therapy. During effective antiviral treatment, the majority of these proviruses remain transcriptionally silent, but mechanisms responsible for viral latency are insufficiently clear. Here, we used matched integration site and proviral sequencing (MIP-Seq), an experimental approach involving multiple displacement amplification of individual proviral species, followed by near-full-length HIV-1 next-generation sequencing and corresponding chromosomal integration site analysis to selectively map the chromosomal positions of intact and defective proviruses in 3 HIV-1-infected individuals undergoing long-term antiretroviral therapy. Simultaneously, chromatin accessibility and gene expression in autologous CD4+ T cells were analyzed by assays for transposase-accessible chromatin using sequencing (ATAC-Seq) and RNA-Seq. We observed that in comparison to proviruses with defective sequences, intact HIV-1 proviruses were enriched for non-genic chromosomal positions and more frequently showed an opposite orientation relative to host genes. In addition, intact HIV-1 proviruses were preferentially integrated in either relative proximity to or increased distance from active transcriptional start sites and to accessible chromatin regions. These studies strongly suggest selection of intact proviruses with features of deeper viral latency during prolonged antiretroviral therapy, and may be informative for targeting the genome-intact viral reservoir.
Collapse
Affiliation(s)
- Kevin B Einkauf
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Guinevere Q Lee
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Radwa Sharaf
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Xiaoming Sun
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Stephane Hua
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Samantha My Chen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Chenyang Jiang
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Xiaodong Lian
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Eric S Rosenberg
- Infectious Disease Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tae-Wook Chun
- National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, Maryland, USA
| | - Jonathan Z Li
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Mathias Lichterfeld
- Infectious Disease Division, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
41
|
Hamada M, Nishio N, Okuno Y, Suzuki S, Kawashima N, Muramatsu H, Tsubota S, Wilson MH, Morita D, Kataoka S, Ichikawa D, Murakami N, Taniguchi R, Suzuki K, Kojima D, Sekiya Y, Nishikawa E, Narita A, Hama A, Kojima S, Nakazawa Y, Takahashi Y. Integration Mapping of piggyBac-Mediated CD19 Chimeric Antigen Receptor T Cells Analyzed by Novel Tagmentation-Assisted PCR. EBioMedicine 2018; 34:18-26. [PMID: 30082227 PMCID: PMC6116345 DOI: 10.1016/j.ebiom.2018.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/28/2018] [Accepted: 07/09/2018] [Indexed: 01/09/2023] Open
Abstract
Insertional mutagenesis is an important risk with all genetically modified cell therapies, including chimeric antigen receptor (CAR)-T cell therapy used for hematological malignancies. Here we describe a new tagmentation-assisted PCR (tag-PCR) system that can determine the integration sites of transgenes without using restriction enzyme digestion (which can potentially bias the detection) and allows library preparation in fewer steps than with other methods. Using this system, we compared the integration sites of CD19-specific CAR genes in final T cell products generated by retrovirus-based and lentivirus-based gene transfer and by the piggyBac transposon system. The piggyBac system demonstrated lower preference than the retroviral system for integration near transcriptional start sites and CpG islands and higher preference than the lentiviral system for integration into genomic safe harbors. Integration into or near proto-oncogenes was similar in all three systems. Tag-PCR mapping is a useful technique for assessing the risk of insertional mutagenesis.
Collapse
Affiliation(s)
- Motoharu Hamada
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhiro Nishio
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Yusuke Okuno
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Satoshi Suzuki
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Nozomu Kawashima
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoma Tsubota
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Matthew H Wilson
- Vanderbilt University School of Medicine, Nashville, TN 37232, United States; VA Tennessee Valley Health Care, Nashville, TN 37212, United States
| | - Daisuke Morita
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shinsuke Kataoka
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Ichikawa
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norihiro Murakami
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rieko Taniguchi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kyogo Suzuki
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daiei Kojima
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuko Sekiya
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Eri Nishikawa
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Narita
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Asahito Hama
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiji Kojima
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
42
|
Biasco L. Integration Site Analysis in Gene Therapy Patients: Expectations and Reality. Hum Gene Ther 2018; 28:1122-1129. [PMID: 29160103 DOI: 10.1089/hum.2017.183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Integration site (IS) analysis is one of the major tools for addressing the safety of gene therapy clinical protocols based on the use of integrating vectors. Over the past years, the study of viral insertions in gene therapy-treated patients has allowed identifying insertional mutagenesis events, evaluating the safety of new viral vector platforms and tracking the in vivo clonal dynamics of genetically engineered cell products. While gene therapy is progressively expanding its impact on a broader area of clinical applications, increasingly more accessible, faster, and more reliable safety readouts are required from IS analysis. Several actors, from researchers to clinicians, from regulatory agencies to private companies, have to interface to different degrees with the results of IS analysis while developing and evaluating gene therapy products based on retroviral vectors. This review is aimed at providing a brief overview of what the current state and the future is of these studies with a particular focus on what are the main analytical constraints that should be considered upon conducting IS analysis in clinical gene therapy.
Collapse
Affiliation(s)
- Luca Biasco
- 1 Harvard Medical School, Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts.,2 University College London , Great Ormond Street Institute of Child Health, Faculty of Population Health Sciences, London, United Kingdom
| |
Collapse
|
43
|
Spontaneous reactivation of latent HIV-1 promoters is linked to the cell cycle as revealed by a genetic-insulators-containing dual-fluorescence HIV-1-based vector. Sci Rep 2018; 8:10204. [PMID: 29977044 PMCID: PMC6033903 DOI: 10.1038/s41598-018-28161-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 06/05/2018] [Indexed: 12/31/2022] Open
Abstract
Long-lived latently HIV-1-infected cells represent a barrier to cure. We developed a dual-fluorescence HIV-1-based vector containing a pair of genetic insulators flanking a constitutive fluorescent reporter gene to study HIV-1 latency. The protective effects of these genetic insulators are demonstrated through long-term (up to 394 days) stable fluorescence profiles in transduced SUP-T1 cells. Analysis of 1,941 vector integration sites confirmed reproduction of HIV-1 integration patterns. We sorted monoclonal cells representing latent HIV-1 infections and found that both vector integration sites and integrity of the vector genomes influence the reactivation potentials of latent HIV-1 promoters. Interestingly, some latent monoclonal cells exhibited a small cell subpopulation with a spontaneously reactivated HIV-1 promoter. Higher expression levels of genes involved in cell cycle progression are observed in these cell subpopulations compared to their counterparts with HIV-1 promoters that remained latent. Consistently, larger fractions of spontaneously reactivated cells are in the S and G2 phases of the cell cycle. Furthermore, genistein and nocodazole treatments of these cell clones, which halted cells in the G2 phase, resulted in a 1.4–2.9-fold increase in spontaneous reactivation. Taken together, our HIV-1 latency model reveals that the spontaneous reactivation of latent HIV-1 promoters is linked to the cell cycle.
Collapse
|
44
|
Krastev DB, Pettitt SJ, Campbell J, Song F, Tanos BE, Stoynov SS, Ashworth A, Lord CJ. Coupling bimolecular PARylation biosensors with genetic screens to identify PARylation targets. Nat Commun 2018; 9:2016. [PMID: 29789535 PMCID: PMC5964205 DOI: 10.1038/s41467-018-04466-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/01/2018] [Indexed: 12/26/2022] Open
Abstract
Poly (ADP-ribose)ylation is a dynamic protein modification that regulates multiple cellular processes. Here, we describe a system for identifying and characterizing PARylation events that exploits the ability of a PBZ (PAR-binding zinc finger) protein domain to bind PAR with high-affinity. By linking PBZ domains to bimolecular fluorescent complementation biosensors, we developed fluorescent PAR biosensors that allow the detection of temporal and spatial PARylation events in live cells. Exploiting transposon-mediated recombination, we integrate the PAR biosensor en masse into thousands of protein coding genes in living cells. Using these PAR-biosensor "tagged" cells in a genetic screen we carry out a large-scale identification of PARylation targets. This identifies CTIF (CBP80/CBP20-dependent translation initiation factor) as a novel PARylation target of the tankyrase enzymes in the centrosomal region of cells, which plays a role in the distribution of the centrosomal satellites.
Collapse
Affiliation(s)
- Dragomir B Krastev
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Stephen J Pettitt
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - James Campbell
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Feifei Song
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Barbara E Tanos
- The Cancer Therapeutics Division, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Stoyno S Stoynov
- The Institute of Molecular Biology, Bulgarian Academy of Sciences, 1113, Sofia, Bulgaria
| | - Alan Ashworth
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
- UCSF Helen Diller Family Comprehensive Cancer Center, 1450 3rd Street, San Francisco, CA, 94158, USA.
| | - Christopher J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
| |
Collapse
|
45
|
Ball CR, Oppel F, Ehrenberg KR, Dubash TD, Dieter SM, Hoffmann CM, Abel U, Herbst F, Koch M, Werner J, Bergmann F, Ishaque N, Schmidt M, von Kalle C, Scholl C, Fröhling S, Brors B, Weichert W, Weitz J, Glimm H. Succession of transiently active tumor-initiating cell clones in human pancreatic cancer xenografts. EMBO Mol Med 2018; 9:918-932. [PMID: 28526679 PMCID: PMC5494525 DOI: 10.15252/emmm.201607354] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Although tumor-initiating cell (TIC) self-renewal has been postulated to be essential in progression and metastasis formation of human pancreatic adenocarcinoma (PDAC), clonal dynamics of TICs within PDAC tumors are yet unknown. Here, we show that long-term progression of PDAC in serial xenotransplantation is driven by a succession of transiently active TICs producing tumor cells in temporally restricted bursts. Clonal tracking of individual, genetically marked TICs revealed that individual tumors are generated by distinct sets of TICs with very little overlap between subsequent xenograft generations. An unexpected functional and phenotypic plasticity of pancreatic TICs in vivo underlies the recruitment of inactive TIC clones in serial xenografts. The observed clonal succession of TIC activity in serial xenotransplantation is in stark contrast to the continuous activity of limited numbers of self-renewing TICs within a fixed cellular hierarchy observed in other epithelial cancers and emphasizes the need to target TIC activation, rather than a fixed TIC population, in PDAC.
Collapse
Affiliation(s)
- Claudia R Ball
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Oppel
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karl Roland Ehrenberg
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Taronish D Dubash
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian M Dieter
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany
| | - Christopher M Hoffmann
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrich Abel
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Friederike Herbst
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Moritz Koch
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Dresden, Dresden, Germany
| | - Jens Werner
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Surgery, University of Munich, Munich, Germany
| | - Frank Bergmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Naveed Ishaque
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Center for Personalized Oncology, DKFZ-HIPO, DKFZ, Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany
| | - Claudia Scholl
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany.,Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Brors
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wilko Weichert
- German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Weitz
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Dresden, Dresden, Germany
| | - Hanno Glimm
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany .,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
46
|
Wang W, Bartholomae CC, Gabriel R, Deichmann A, Schmidt M. The LAM-PCR Method to Sequence LV Integration Sites. Methods Mol Biol 2018; 1448:107-20. [PMID: 27317177 DOI: 10.1007/978-1-4939-3753-0_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Integrating viral gene transfer vectors are commonly used gene delivery tools in clinical gene therapy trials providing stable integration and continuous gene expression of the transgene in the treated host cell. However, integration of the reverse-transcribed vector DNA into the host genome is a potentially mutagenic event that may directly contribute to unwanted side effects. A comprehensive and accurate analysis of the integration site (IS) repertoire is indispensable to study clonality in transduced cells obtained from patients undergoing gene therapy and to identify potential in vivo selection of affected cell clones. To date, next-generation sequencing (NGS) of vector-genome junctions allows sophisticated studies on the integration repertoire in vitro and in vivo. We have explored the use of the Illumina MiSeq Personal Sequencer platform to sequence vector ISs amplified by non-restrictive linear amplification-mediated PCR (nrLAM-PCR) and LAM-PCR. MiSeq-based high-quality IS sequence retrieval is accomplished by the introduction of a double-barcode strategy that substantially minimizes the frequency of IS sequence collisions compared to the conventionally used single-barcode protocol. Here, we present an updated protocol of (nr)LAM-PCR for the analysis of lentiviral IS using a double-barcode system and followed by deep sequencing using the MiSeq device.
Collapse
Affiliation(s)
- Wei Wang
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Cynthia C Bartholomae
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Richard Gabriel
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Annette Deichmann
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases and German Cancer Research Center, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany.
| |
Collapse
|
47
|
Juanes JM, Gallego A, Tárraga J, Chaves FJ, Marín-Garcia P, Medina I, Arnau V, Dopazo J. VISMapper: ultra-fast exhaustive cartography of viral insertion sites for gene therapy. BMC Bioinformatics 2017; 18:421. [PMID: 28931371 PMCID: PMC5607581 DOI: 10.1186/s12859-017-1837-z] [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: 02/13/2017] [Accepted: 09/12/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The possibility of integrating viral vectors to become a persistent part of the host genome makes them a crucial element of clinical gene therapy. However, viral integration has associated risks, such as the unintentional activation of oncogenes that can result in cancer. Therefore, the analysis of integration sites of retroviral vectors is a crucial step in developing safer vectors for therapeutic use. RESULTS Here we present VISMapper, a vector integration site analysis web server, to analyze next-generation sequencing data for retroviral vector integration sites. VISMapper can be found at: http://vismapper.babelomics.org . CONCLUSIONS Because it uses novel mapping algorithms VISMapper is remarkably faster than previous available programs. It also provides a useful graphical interface to analyze the integration sites found in the genomic context.
Collapse
Affiliation(s)
- José M Juanes
- Departamento de Informática, Escuela Técnica Superior de Ingeniería (ETSE), Universidad de Valencia, 46100, Valencia, Burjassot, Spain.,Computational Genomics Department, Prince Felipe Research Center, 46012, Valencia, Spain
| | - Asunción Gallego
- Clinical Bioinformatics Research Area, Fundación Progreso y Salud, Hospital Virgen del Rocío, 41013, Sevilla, Spain.,Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Hospital Virgen del Rocío, 41013, Sevilla, Spain
| | - Joaquín Tárraga
- Computational Genomics Department, Prince Felipe Research Center, 46012, Valencia, Spain.,HPC Service, University Information Services, University of Cambridge, Cambridge, UK
| | - Felipe J Chaves
- Genotyping and Genetic Diagnosis Unit, Health Research Institute, INCLIVA, Valencia, Spain.,CIBERDem, Health Institute Carlos III, Madrid, Spain
| | - Pablo Marín-Garcia
- Genotyping and Genetic Diagnosis Unit, Health Research Institute, INCLIVA, Valencia, Spain.,Institute for Integrative Systems Biology (I2SysBio), Universidad de Valencia-CSIC, 46980, Valencia, Paterna, Spain
| | - Ignacio Medina
- HPC Service, University Information Services, University of Cambridge, Cambridge, UK
| | - Vicente Arnau
- Departamento de Informática, Escuela Técnica Superior de Ingeniería (ETSE), Universidad de Valencia, 46100, Valencia, Burjassot, Spain.,Computational Genomics Department, Prince Felipe Research Center, 46012, Valencia, Spain.,Institute for Integrative Systems Biology (I2SysBio), Universidad de Valencia-CSIC, 46980, Valencia, Paterna, Spain
| | - Joaquín Dopazo
- Clinical Bioinformatics Research Area, Fundación Progreso y Salud, Hospital Virgen del Rocío, 41013, Sevilla, Spain. .,Bioinformatics and Data Analysis Unit, Genomic Medicine Institute Imegen, Valencia, Spain. .,Functional Genomics Node, INB-ELIXIR-es, Hospital Virgen del Rocío, 42013, Sevilla, Spain.
| |
Collapse
|
48
|
Scholz SJ, Fronza R, Bartholomä CC, Cesana D, Montini E, von Kalle C, Gil-Farina I, Schmidt M. Lentiviral Vector Promoter is Decisive for Aberrant Transcript Formation. Hum Gene Ther 2017; 28:875-885. [PMID: 28825370 DOI: 10.1089/hum.2017.162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Lentiviral vectors hold great promise for the genetic correction of various inherited diseases. However, lentiviral vector biology is still not completely understood and warrants the precise decoding of molecular mechanisms underlying integration and post-translational modification. This study investigated a series of self-inactivating (SIN) and full long terminal repeat (LTR) lentiviral vectors that contained different types of promoters with or without a transgene to gain deeper insights in lentiviral target site selection and potential perturbation of cellular gene expression. Using an optimized nonrestrictive linear amplification-mediated polymerase chain reaction (nrLAM-PCR) protocol, vector structure-dependent integration site profiles were observed upon transduction of mouse lin- hematopoietic progenitors in vitro. Initial target site selection mainly depended on the presence of the promoter while being independent of its nature. Despite the increased propensity for read-through transcription of SIN lentiviral vectors, the incidence of viral-cellular fusion transcript formation involving the canonical viral splice donor or cryptic splice sites was reduced in both unselected primary lin- cells and transformed 32D cells. Moreover, the strength of the internal promoter in vectors with SIN LTRs is decisive for in vitro selection and for the abundance of chimeric transcripts, which are decreased by moderately active promoters. These results will help to better understand vector biology and to optimize therapeutic vectors for future gene therapy applications.
Collapse
Affiliation(s)
- Simone J Scholz
- 1 Department of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases , Heidelberg, Germany
| | - Raffaele Fronza
- 1 Department of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases , Heidelberg, Germany .,2 GeneWerk GmbH, Heidelberg, Germany
| | - Cynthia C Bartholomä
- 1 Department of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases , Heidelberg, Germany
| | - Daniela Cesana
- 3 San Raffaele Telethon Institute for Gene Therapy , Milan, Italy
| | - Eugenio Montini
- 3 San Raffaele Telethon Institute for Gene Therapy , Milan, Italy
| | - Christof von Kalle
- 1 Department of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases , Heidelberg, Germany
| | - Irene Gil-Farina
- 1 Department of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases , Heidelberg, Germany
| | - Manfred Schmidt
- 1 Department of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases , Heidelberg, Germany .,2 GeneWerk GmbH, Heidelberg, Germany
| |
Collapse
|
49
|
Spaller T, Groth M, Glöckner G, Winckler T. TRE5-A retrotransposition profiling reveals putative RNA polymerase III transcription complex binding sites on the Dictyostelium extrachromosomal rDNA element. PLoS One 2017; 12:e0175729. [PMID: 28406973 PMCID: PMC5391098 DOI: 10.1371/journal.pone.0175729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/30/2017] [Indexed: 11/28/2022] Open
Abstract
The amoeba Dictyostelium discoideum has a haploid genome in which two thirds of the DNA encodes proteins. Consequently, the space available for selfish mobile elements to expand without excess damage to the host genome is limited. The non-long terminal repeat retrotransposon TRE5-A maintains an active population in the D. discoideum genome and apparently adapted to this gene-dense environment by targeting positions ~47 bp upstream of tRNA genes that are devoid of protein-coding regions. Because only ~24% of tRNA genes are associated with a TRE5-A element in the reference genome, we evaluated whether TRE5-A retrotransposition is limited to this subset of tRNA genes. We determined that a tagged TRE5-A element (TRE5-Absr) integrated at 384 of 405 tRNA genes, suggesting that expansion of the current natural TRE5-A population is not limited by the availability of targets. We further observed that TRE5-Absr targets the ribosomal 5S gene on the multicopy extrachromosomal DNA element that carries the ribosomal RNA genes, indicating that TRE5-A integration may extend to the entire RNA polymerase III (Pol III) transcriptome. We determined that both natural TRE5-A and cloned TRE5-Absr retrotranspose to locations on the extrachromosomal rDNA element that contain tRNA gene-typical A/B box promoter motifs without displaying any other tRNA gene context. Based on previous data suggesting that TRE5-A targets tRNA genes by locating Pol III transcription complexes, we propose that A/B box loci reflect Pol III transcription complex assembly sites that possess a function in the biology of the extrachromosomal rDNA element.
Collapse
MESH Headings
- Binding Sites
- DNA, Protozoan/chemistry
- DNA, Protozoan/genetics
- DNA, Protozoan/metabolism
- DNA, Ribosomal/chemistry
- DNA, Ribosomal/genetics
- DNA, Ribosomal/metabolism
- Dictyostelium/genetics
- Promoter Regions, Genetic
- Protozoan Proteins/metabolism
- RNA Polymerase III/metabolism
- RNA, Ribosomal, 5S/genetics
- RNA, Transfer/genetics
- Retroelements/genetics
- Transcription, Genetic
Collapse
Affiliation(s)
- Thomas Spaller
- Pharmaceutical Biology, Institute of Pharmacy, University of Jena, Jena, Germany
| | - Marco Groth
- Core Facility DNA Sequencing, Leibniz Institute for Age Research–Fritz Lipmann Institute, Jena, Germany
| | - Gernot Glöckner
- Institute for Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
| | - Thomas Winckler
- Pharmaceutical Biology, Institute of Pharmacy, University of Jena, Jena, Germany
- * E-mail:
| |
Collapse
|
50
|
Cytoreductive conditioning intensity predicts clonal diversity in ADA-SCID retroviral gene therapy patients. Blood 2017; 129:2624-2635. [PMID: 28351939 DOI: 10.1182/blood-2016-12-756734] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/20/2017] [Indexed: 01/15/2023] Open
Abstract
Retroviral gene therapy has proved efficacious for multiple genetic diseases of the hematopoietic system, but roughly half of clinical gene therapy trial protocols using gammaretroviral vectors have reported leukemias in some of the patients treated. In dramatic contrast, 39 adenosine deaminase-deficient severe combined immunodeficiency (ADA-SCID) patients have been treated with 4 distinct gammaretroviral vectors without oncogenic consequence. We investigated clonal dynamics and diversity in a cohort of 15 ADA-SCID children treated with gammaretroviral vectors and found clear evidence of genotoxicity, indicated by numerous common integration sites near proto-oncogenes and by increased abundance of clones with integrations near MECOM and LMO2 These clones showed stable behavior over multiple years and never expanded to the point of dominance or dysplasia. One patient developed a benign clonal dominance that could not be attributed to insertional mutagenesis and instead likely resulted from expansion of a transduced natural killer clone in response to chronic Epstein-Barr virus viremia. Clonal diversity and T-cell repertoire, measured by vector integration site sequencing and T-cell receptor β-chain rearrangement sequencing, correlated significantly with the amount of busulfan preconditioning delivered to patients and to CD34+ cell dose. These data, in combination with results of other ADA-SCID gene therapy trials, suggest that disease background may be a crucial factor in leukemogenic potential of retroviral gene therapy and underscore the importance of cytoreductive conditioning in this type of gene therapy approach.
Collapse
|