Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
BMC Bioinformatics ; 24(1): 286, 2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37464281

ABSTRACT

BACKGROUND: Integration site (IS) analysis is a fundamental analytical platform for evaluating the safety and efficacy of viral vector based preclinical and clinical Gene Therapy (GT). A handful of groups have developed standardized bioinformatics pipelines to process IS sequencing data, to generate reports, and/or to perform comparative studies across different GT trials. Keeping up with the technological advances in the field of IS analysis, different computational pipelines have been published over the past decade. These pipelines focus on identifying IS from single-read sequencing or paired-end sequencing data either using read-based or using sonication fragment-based methods, but there is a lack of a bioinformatics tool that automatically includes unique molecular identifiers (UMI) for IS abundance estimations and allows comparing multiple quantification methods in one integrated pipeline. RESULTS: Here we present IS-Seq a bioinformatics pipeline that can process data from paired-end sequencing of both old restriction sites-based IS collection methods and new sonication-based IS retrieval systems while allowing the selection of different abundance estimation methods, including read-based, Fragment-based and UMI-based systems. CONCLUSIONS: We validated the performance of IS-Seq by testing it against the most popular  analytical workflow available in the literature (INSPIIRED) and using different scenarios. Lastly, by performing extensive simulation studies and a comprehensive wet-lab assessment of our IS-Seq pipeline we could show that in clinically relevant scenarios, UMI quantification provides better accuracy than the currently most widely used sonication fragment counts as a method for IS abundance estimation.


Subject(s)
Computational Biology , High-Throughput Nucleotide Sequencing , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA , Genetic Vectors
2.
Mol Ther ; 30(10): 3209-3225, 2022 10 05.
Article in English | MEDLINE | ID: mdl-35614857

ABSTRACT

Hematopoietic stem/progenitor cell gene therapy (HSPC-GT) has shown clear neurological benefit in rare diseases, which is achieved through the engraftment of genetically modified microglia-like cells (MLCs) in the brain. Still, the engraftment dynamics and the nature of engineered MLCs, as well as their potential use in common neurogenerative diseases, have remained largely unexplored. Here, we comprehensively characterized how different routes of administration affect the biodistribution of genetically engineered MLCs and other HSPC derivatives in mice. We generated a high-resolution single-cell transcriptional map of MLCs and discovered that they could clearly be distinguished from macrophages as well as from resident microglia by the expression of a specific gene signature that is reflective of their HSPC ontogeny and irrespective of their long-term engraftment history. Lastly, using murine models of Parkinson's disease and frontotemporal dementia, we demonstrated that MLCs can deliver therapeutically relevant levels of transgenic protein to the brain, thereby opening avenues for the clinical translation of HSPC-GT to the treatment of major neurological diseases.


Subject(s)
Hematopoietic Stem Cell Transplantation , Animals , Genetic Engineering , Genetic Therapy , Hematopoietic Stem Cells/metabolism , Mice , Tissue Distribution
3.
Mol Ther ; 20(4): 709-16, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22252453

ABSTRACT

γ-Retroviral and lentiviral vectors allow the permanent integration of a therapeutic transgene in target cells and have provided in the last decade a delivery platform for several successful gene therapy (GT) clinical approaches. However, the occurrence of adverse events due to insertional mutagenesis in GT treated patients poses a strong challenge to the scientific community to identify the mechanisms at the basis of vector-driven genotoxicity. Along the last decade, the study of retroviral integration sites became a fundamental tool to monitor vector-host interaction in patients overtime. This review is aimed at critically revising the data derived from insertional profiling, with a particular focus on the evidences collected from GT clinical trials. We discuss the controversies and open issues associated to the interpretation of integration site analysis during patient's follow up, with an update on the latest results derived from the use of high-throughput technologies. Finally, we provide a perspective on the future technical development and on the application of these studies to address broader biological questions, from basic virology to human hematopoiesis.


Subject(s)
Genetic Therapy/methods , Retroviridae/genetics , Genetic Therapy/adverse effects , Humans , Mutagenesis, Insertional/methods
4.
Nat Commun ; 12(1): 1622, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712608

ABSTRACT

Our mathematical model of integration site data in clinical gene therapy supported the existence of long-term lymphoid progenitors capable of surviving independently from hematopoietic stem cells. To date, no experimental setting has been available to validate this prediction. We here report evidence of a population of lymphoid progenitors capable of independently maintaining T and NK cell production for 15 years in humans. The gene therapy patients of this study lack vector-positive myeloid/B cells indicating absence of engineered stem cells but retain gene marking in both T and NK. Decades after treatment, we can still detect and analyse transduced naïve T cells whose production is likely maintained by a population of long-term lymphoid progenitors. By tracking insertional clonal markers overtime, we suggest that these progenitors can support both T and NK cell production. Identification of these long-term lymphoid progenitors could be utilised for the development of next generation gene- and cancer-immunotherapies.


Subject(s)
Killer Cells, Natural/physiology , Lymphocytes/physiology , Lymphoid Progenitor Cells/physiology , T-Lymphocytes/physiology , B-Lymphocytes , Genetic Therapy/methods , Hematopoietic Stem Cells , Humans , Interferon-gamma/metabolism , Mutagenesis , Myeloid Cells/physiology , Proto-Oncogenes/genetics , Proto-Oncogenes/physiology
5.
J Cell Commun Signal ; 14(3): 335-347, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32504411

ABSTRACT

Appropriate tools for monitoring sarcoma progression are still limited. The aim of the present study was to investigate the value of miR-34a-5p (miR34a) as a circulating biomarker to follow disease progression and measure the therapeutic response. Stable forced re-expression of miR34a in Ewing sarcoma (EWS) cells significantly limited tumor growth in mice. Absolute quantification of miR34a in the plasma of mice and 31 patients showed that high levels of this miRNA inversely correlated with tumor volume. In addition, miR34a expression was higher in the blood of localized EWS patients than in the blood of metastatic EWS patients. In 12 patients, we followed miR34a expression during preoperative chemotherapy. While there was no variation in the blood miR34a levels in metastatic patients at the time of diagnosis or after the last cycle of preoperative chemotherapy, there was an increase in the circulating miR34a levels in patients with localized tumors. The three patients with the highest fold-increase in the miR levels did not show evidence of metastasis. Although this analysis should be extended to a larger cohort of patients, these findings imply that detection of the miR34a levels in the blood of EWS patients may assist with the clinical management of EWS.

6.
Nat Commun ; 10(1): 2395, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160568

ABSTRACT

Hematopoietic Stem/Progenitor cells (HSPCs) are endowed with the role of maintaining a diverse pool of blood cells throughout the human life. Despite recent efforts, the nature of the early cell fate decisions remains contentious. Using single-cell RNA-Seq, we show that existing approaches to stratify bone marrow CD34+ cells reveal a hierarchically-structured transcriptional landscape of hematopoietic differentiation. Still, this landscape misses important early fate decisions. We here provide a broader transcriptional profiling of bone marrow lineage negative hematopoietic progenitors that recovers a key missing branchpoint into basophils and expands our understanding of the underlying structure of early adult human haematopoiesis. We also show that this map has strong similarities in topology and gene expression to that found in mouse. Finally, we identify the sialomucin CD164, as a reliable marker for the earliest branches of HSPCs specification and we showed how its use can foster the design of alternative transplantation cell products.


Subject(s)
Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Animals , Antigens, CD34/metabolism , Bone Marrow Cells , Cell Lineage , Endolyn/metabolism , Gene Expression Profiling , Humans , Mice , Sequence Analysis, RNA , Single-Cell Analysis
7.
Nat Med ; 25(5): 776-783, 2019 05.
Article in English | MEDLINE | ID: mdl-30911135

ABSTRACT

Re-expression of the paralogous γ-globin genes (HBG1/2) could be a universal strategy to ameliorate the severe ß-globin disorders sickle cell disease (SCD) and ß-thalassemia by induction of fetal hemoglobin (HbF, α2γ2)1. Previously, we and others have shown that core sequences at the BCL11A erythroid enhancer are required for repression of HbF in adult-stage erythroid cells but are dispensable in non-erythroid cells2-6. CRISPR-Cas9-mediated gene modification has demonstrated variable efficiency, specificity, and persistence in hematopoietic stem cells (HSCs). Here, we demonstrate that Cas9:sgRNA ribonucleoprotein (RNP)-mediated cleavage within a GATA1 binding site at the +58 BCL11A erythroid enhancer results in highly penetrant disruption of this motif, reduction of BCL11A expression, and induction of fetal γ-globin. We optimize conditions for selection-free on-target editing in patient-derived HSCs as a nearly complete reaction lacking detectable genotoxicity or deleterious impact on stem cell function. HSCs preferentially undergo non-homologous compared with microhomology-mediated end joining repair. Erythroid progeny of edited engrafting SCD HSCs express therapeutic levels of HbF and resist sickling, while those from patients with ß-thalassemia show restored globin chain balance. Non-homologous end joining repair-based BCL11A enhancer editing approaching complete allelic disruption in HSCs is a practicable therapeutic strategy to produce durable HbF induction.


Subject(s)
Gene Editing/methods , Hematopoietic Stem Cells/metabolism , Amino Acid Sequence , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Base Sequence , CRISPR-Cas Systems , Carrier Proteins/genetics , Enhancer Elements, Genetic , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Hematopoietic Stem Cell Transplantation , Humans , INDEL Mutation , Nuclear Proteins/genetics , RNA, Guide, Kinetoplastida/genetics , Repressor Proteins , beta-Thalassemia/blood , beta-Thalassemia/genetics , beta-Thalassemia/therapy , gamma-Globins/biosynthesis , gamma-Globins/genetics
8.
Blood Adv ; 2(19): 2505-2512, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30282642

ABSTRACT

Novel therapies for sickle cell disease (SCD) based on genetically engineered autologous hematopoietic stem and progenitor cells (HSPCs) are critically dependent on a safe and effective strategy for cell procurement. We sought to assess the safety and efficacy of plerixafor when used in transfused patients with SCD for HSC mobilization. Six adult patients with SCD were recruited to receive a single dose of plerixafor, tested at lower than standard (180 µg/kg) and standard (240 µg/kg) doses, followed by CD34+ cell monitoring in peripheral blood and apheresis collection. The procedures were safe and well-tolerated. Mobilization was successful, with higher peripheral CD34+ cell counts in the standard vs the low-dose group. Among our 6 donors, we improved apheresis cell collection results by using a deep collection interface and starting apheresis within 4 hours after plerixafor administration. In the subjects who received a single standard dose of plerixafor and followed the optimized collection protocol, yields of up to 24.5 × 106 CD34+ cells/kg were achieved. Interestingly, the collected CD34+ cells were enriched in immunophenotypically defined long-term HSCs and early progenitors. Thus, we demonstrate that plerixafor can be employed safely in patients with SCD to obtain sufficient HSCs for potential use in gene therapy.


Subject(s)
Anemia, Sickle Cell/therapy , Blood Component Removal , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/administration & dosage , Adolescent , Adult , Benzylamines , Cyclams , Dose-Response Relationship, Drug , Genetic Therapy/methods , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Peripheral Blood Stem Cell Transplantation/methods , Pilot Projects , Young Adult
9.
Cell Stem Cell ; 19(1): 107-19, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27237736

ABSTRACT

Hematopoietic stem/progenitor cells (HSPCs) are capable of supporting the lifelong production of blood cells exerting a wide spectrum of functions. Lentiviral vector HSPC gene therapy generates a human hematopoietic system stably marked at the clonal level by vector integration sites (ISs). Using IS analysis, we longitudinally tracked >89,000 clones from 15 distinct bone marrow and peripheral blood lineages purified up to 4 years after transplant in four Wiskott-Aldrich syndrome patients treated with HSPC gene therapy. We measured at the clonal level repopulating waves, populations' sizes and dynamics, activity of distinct HSPC subtypes, contribution of various progenitor classes during the early and late post-transplant phases, and hierarchical relationships among lineages. We discovered that in-vitro-manipulated HSPCs retain the ability to return to latency after transplant and can be physiologically reactivated, sustaining a stable hematopoietic output. This study constitutes in vivo comprehensive tracking in humans of hematopoietic clonal dynamics during the early and late post-transplant phases.


Subject(s)
Cell Tracking , Hematopoiesis , Antigens, CD34/metabolism , Cell Engineering , Cell Lineage/genetics , Child, Preschool , Clone Cells , Genetic Therapy , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Infant , Male , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Mutagenesis, Insertional/genetics , Time Factors , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/therapy
10.
Sci Transl Med ; 7(273): 273ra13, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25653219

ABSTRACT

A definitive understanding of survival and differentiation potential in humans of T cell subpopulations is of paramount importance for the development of effective T cell therapies. In particular, uncovering the dynamics in vivo in humans of the recently described T memory stem cells (TSCM) would be crucial for therapeutic approaches that aim at taking advantage of a stable cellular vehicle with precursor potential. We exploited data derived from two gene therapy clinical trials for an inherited immunodeficiency, using either retrovirally engineered hematopoietic stem cells or mature lymphocytes to trace individual T cell clones directly in vivo in humans. We compared healthy donors and bone marrow-transplanted patients, studied long-term in vivo T cell composition under different clinical conditions, and specifically examined TSCM contribution according to age, conditioning regimen, disease background, cell source, long-term reconstitution, and ex vivo gene correction processing. High-throughput sequencing of retroviral vector integration sites (ISs) allowed tracing the fate of more than 1700 individual T cell clones in gene therapy patients after infusion of gene-corrected hematopoietic stem cells or mature lymphocytes. We shed light on long-term in vivo clonal relationships among different T cell subtypes, and we unveiled that TSCM are able to persist and to preserve their precursor potential in humans for up to 12 years after infusion of gene-corrected lymphocytes. Overall, this work provides high-resolution tracking of T cell fate and activity and validates, in humans, the safe and functional decade-long survival of engineered TSCM, paving the way for their future application in clinical settings.


Subject(s)
Cell Tracking , Genetic Engineering , Hematopoietic Stem Cells/cytology , Immunologic Memory , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Adult , Cell Survival , Child , Clone Cells , Genetic Therapy , Humans , Interleukin-2/pharmacology , Longitudinal Studies , Lymphocyte Subsets/metabolism , Phenotype , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Tissue Donors
11.
Science ; 341(6148): 1233151, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23845947

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is an inherited immunodeficiency caused by mutations in the gene encoding WASP, a protein regulating the cytoskeleton. Hematopoietic stem/progenitor cell (HSPC) transplants can be curative, but, when matched donors are unavailable, infusion of autologous HSPCs modified ex vivo by gene therapy is an alternative approach. We used a lentiviral vector encoding functional WASP to genetically correct HSPCs from three WAS patients and reinfused the cells after a reduced-intensity conditioning regimen. All three patients showed stable engraftment of WASP-expressing cells and improvements in platelet counts, immune functions, and clinical scores. Vector integration analyses revealed highly polyclonal and multilineage haematopoiesis resulting from the gene-corrected HSPCs. Lentiviral gene therapy did not induce selection of integrations near oncogenes, and no aberrant clonal expansion was observed after 20 to 32 months. Although extended clinical observation is required to establish long-term safety, lentiviral gene therapy represents a promising treatment for WAS.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/therapy , Child , Genetic Vectors , Humans , Lentivirus , Male , Transduction, Genetic , Virus Integration
SELECTION OF CITATIONS
SEARCH DETAIL