Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30
Filtrer
1.
Methods Mol Biol ; 1448: 107-20, 2016.
Article de Anglais | MEDLINE | ID: mdl-27317177

RÉSUMÉ

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.


Sujet(s)
Techniques de transfert de gènes , Lentivirus/génétique , Intégration virale/génétique , Thérapie génétique/méthodes , Vecteurs génétiques , Séquençage nucléotidique à haut débit/méthodes , Humains , Réaction de polymérisation en chaîne/méthodes
2.
Sci Transl Med ; 7(277): 277ra28, 2015 Mar 04.
Article de Anglais | MEDLINE | ID: mdl-25739762

RÉSUMÉ

We investigated the efficacy of liver-directed gene therapy using lentiviral vectors in a large animal model of hemophilia B and evaluated the risk of insertional mutagenesis in tumor-prone mouse models. We showed that gene therapy using lentiviral vectors targeting the expression of a canine factor IX transgene in hepatocytes was well tolerated and provided a stable long-term production of coagulation factor IX in dogs with hemophilia B. By exploiting three different mouse models designed to amplify the consequences of insertional mutagenesis, we showed that no genotoxicity was detected with these lentiviral vectors. Our findings suggest that lentiviral vectors may be an attractive candidate for gene therapy targeted to the liver and may be potentially useful for the treatment of hemophilia.


Sujet(s)
Thérapie génétique , Hémophilie B/thérapie , Lentivirus/génétique , Foie/anatomopathologie , Animaux , Coagulation sanguine , Modèles animaux de maladie humaine , Chiens , Femelle , Vecteurs génétiques/métabolisme , Souris de lignée C57BL , Mutagènes/toxicité , Facteurs temps , Transduction génétique , Transgènes
3.
J Vis Exp ; (88): e51543, 2014 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-24998871

RÉSUMÉ

Linear-amplification mediated PCR (LAM-PCR) has been developed to study hematopoiesis in gene corrected cells of patients treated by gene therapy with integrating vector systems. Due to the stable integration of retroviral vectors, integration sites can be used to study the clonal fate of individual cells and their progeny. LAM- PCR for the first time provided evidence that leukemia in gene therapy treated patients originated from provirus induced overexpression of a neighboring proto-oncogene. The high sensitivity and specificity of LAM-PCR compared to existing methods like inverse PCR and ligation mediated (LM)-PCR is achieved by an initial preamplification step (linear PCR of 100 cycles) using biotinylated vector specific primers which allow subsequent reaction steps to be carried out on solid phase (magnetic beads). LAM-PCR is currently the most sensitive method available to identify unknown DNA which is located in the proximity of known DNA. Recently, a variant of LAM-PCR has been developed that circumvents restriction digest thus abrogating retrieval bias of integration sites and enables a comprehensive analysis of provirus locations in host genomes. The following protocol explains step-by-step the amplification of both 3'- and 5'- sequences adjacent to the integrated lentiviral vector.


Sujet(s)
ADN/analyse , Réaction de polymérisation en chaîne/méthodes , ADN/génétique , Vecteurs génétiques/analyse , Vecteurs génétiques/génétique , Séquençage nucléotidique à haut débit/méthodes , Lentivirus/génétique , Retroviridae/génétique
4.
Nature ; 511(7510): 428-34, 2014 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-25043047

RÉSUMÉ

Medulloblastoma is a highly malignant paediatric brain tumour currently treated with a combination of surgery, radiation and chemotherapy, posing a considerable burden of toxicity to the developing child. Genomics has illuminated the extensive intertumoral heterogeneity of medulloblastoma, identifying four distinct molecular subgroups. Group 3 and group 4 subgroup medulloblastomas account for most paediatric cases; yet, oncogenic drivers for these subtypes remain largely unidentified. Here we describe a series of prevalent, highly disparate genomic structural variants, restricted to groups 3 and 4, resulting in specific and mutually exclusive activation of the growth factor independent 1 family proto-oncogenes, GFI1 and GFI1B. Somatic structural variants juxtapose GFI1 or GFI1B coding sequences proximal to active enhancer elements, including super-enhancers, instigating oncogenic activity. Our results, supported by evidence from mouse models, identify GFI1 and GFI1B as prominent medulloblastoma oncogenes and implicate 'enhancer hijacking' as an efficient mechanism driving oncogene activation in a childhood cancer.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Éléments activateurs (génétique)/génétique , Variation structurale du génome/génétique , Médulloblastome/génétique , Oncogènes/génétique , Protéines proto-oncogènes/génétique , Protéines de répression/génétique , Facteurs de transcription/génétique , Animaux , Enfant , Chromosomes humains de la paire 9/génétique , Protéines de liaison à l'ADN/métabolisme , Humains , Médulloblastome/classification , Médulloblastome/anatomopathologie , Souris , Protéines proto-oncogènes/métabolisme , Protéines de répression/métabolisme , Facteurs de transcription/métabolisme
5.
J Antimicrob Chemother ; 69(10): 2809-18, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-24962031

RÉSUMÉ

OBJECTIVES: The rapid early-phase decay of plasma HIV-1 RNA during integrase inhibitor-based therapy is not fully understood. The accumulation of biologically active episomal HIV-1 cDNAs, following aborted integration, could contribute to antiviral potency in vivo. METHODS: This prospective, controlled clinical observation study explored raltegravir's impact on the dynamics of HIV-1 RNA in plasma, and concentrations of total HIV-1 cDNA, episomal 2-long terminal repeat (LTR) circles and HIV-1 integrants in peripheral blood mononuclear cells (PBMC). Individuals starting therapy with two nucleoside reverse transcriptase inhibitors plus either raltegravir (raltegravir group; n = 10 patients) or boosted protease inhibitor/non-nucleoside reverse transcriptase inhibitor (control group; n = 10 patients) were followed for 48 weeks. RESULTS: Suppression of HIV-1 RNA (<50 copies/mL) was reached earlier (5/10 versus 0/10 at week 4; 8/10 versus 4/10 at week 12) on raltegravir. Significant total HIV-1 cDNA reductions in PBMC were reached by day 99 and persisted until day 330, with median factors of decrease of 7.2 and 8.9, respectively. Broad inter-individual variations, yet no treatment-associated differences, were noted for HIV-1 cDNA concentrations. Despite reductions in HIV-1 RNA (∼3 log) and total HIV-1 cDNA (∼1 log), concentrations of integrants and 2-LTR circles remained largely unchanged. CONCLUSIONS: These results extend the previously reported early benefit of raltegravir on the decline of plasma viraemia to treatment-naive patients. The modest treatment-associated, yet group-independent, decline in total HIV-1 cDNA load and the lack of significant changes in integrated and episomal HIV-1 cDNA suggest that most integrated DNA is archival and targeting of HIV reservoirs other than PBMC may underlie beneficial effects of raltegravir.


Sujet(s)
Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , Inhibiteurs de l'intégrase du VIH/pharmacologie , Inhibiteurs de l'intégrase du VIH/usage thérapeutique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Pyrrolidones/pharmacologie , Pyrrolidones/usage thérapeutique , Adulte , Sujet âgé , Femelle , Répétition terminale longue du VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Humains , Agranulocytes/virologie , Mâle , Adulte d'âge moyen , Projets pilotes , Provirus , ARN viral , Raltégravir de potassium , Charge virale , Intégration virale , Jeune adulte
6.
Nature ; 510(7506): 537-41, 2014 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-24847876

RÉSUMÉ

Epigenetic alterations, that is, disruption of DNA methylation and chromatin architecture, are now acknowledged as a universal feature of tumorigenesis. Medulloblastoma, a clinically challenging, malignant childhood brain tumour, is no exception. Despite much progress from recent genomics studies, with recurrent changes identified in each of the four distinct tumour subgroups (WNT-pathway-activated, SHH-pathway-activated, and the less-well-characterized Group 3 and Group 4), many cases still lack an obvious genetic driver. Here we present whole-genome bisulphite-sequencing data from thirty-four human and five murine tumours plus eight human and three murine normal controls, augmented with matched whole-genome, RNA and chromatin immunoprecipitation sequencing data. This comprehensive data set allowed us to decipher several features underlying the interplay between the genome, epigenome and transcriptome, and its effects on medulloblastoma pathophysiology. Most notable were highly prevalent regions of hypomethylation correlating with increased gene expression, extending tens of kilobases downstream of transcription start sites. Focal regions of low methylation linked to transcription-factor-binding sites shed light on differential transcriptional networks between subgroups, whereas increased methylation due to re-normalization of repressed chromatin in DNA methylation valleys was positively correlated with gene expression. Large, partially methylated domains affecting up to one-third of the genome showed increased mutation rates and gene silencing in a subgroup-specific fashion. Epigenetic alterations also affected novel medulloblastoma candidate genes (for example, LIN28B), resulting in alternative promoter usage and/or differential messenger RNA/microRNA expression. Analysis of mouse medulloblastoma and precursor-cell methylation demonstrated a somatic origin for many alterations. Our data provide insights into the epigenetic regulation of transcription and genome organization in medulloblastoma pathogenesis, which are probably also of importance in a wider developmental and disease context.


Sujet(s)
Méthylation de l'ADN/génétique , Régulation de l'expression des gènes tumoraux , Extinction de l'expression des gènes , Médulloblastome/génétique , Analyse de séquence d'ADN/méthodes , Animaux , Sites de fixation , Lignée cellulaire tumorale , Chromatine/génétique , Chromatine/métabolisme , Immunoprécipitation de la chromatine , Femelle , Génome/génétique , Histone/métabolisme , Humains , Médulloblastome/anatomopathologie , Souris , Régions promotrices (génétique)/génétique , Protéines de liaison à l'ARN/génétique , Facteurs de transcription/métabolisme , Transcription génétique
7.
Cancer Cell ; 25(3): 393-405, 2014 Mar 17.
Article de Anglais | MEDLINE | ID: mdl-24651015

RÉSUMÉ

Smoothened (SMO) inhibitors recently entered clinical trials for sonic-hedgehog-driven medulloblastoma (SHH-MB). Clinical response is highly variable. To understand the mechanism(s) of primary resistance and identify pathways cooperating with aberrant SHH signaling, we sequenced and profiled a large cohort of SHH-MBs (n = 133). SHH pathway mutations involved PTCH1 (across all age groups), SUFU (infants, including germline), and SMO (adults). Children >3 years old harbored an excess of downstream MYCN and GLI2 amplifications and frequent TP53 mutations, often in the germline, all of which were rare in infants and adults. Functional assays in different SHH-MB xenograft models demonstrated that SHH-MBs harboring a PTCH1 mutation were responsive to SMO inhibition, whereas tumors harboring an SUFU mutation or MYCN amplification were primarily resistant.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Protéines Hedgehog/génétique , Médulloblastome/génétique , Récepteurs couplés aux protéines G/antagonistes et inhibiteurs , Récepteurs couplés aux protéines G/génétique , Adolescent , Adulte , Animaux , Séquence nucléotidique , Dérivés du biphényle/usage thérapeutique , Tumeurs du cervelet/traitement médicamenteux , Tumeurs du cervelet/génétique , Enfant , Enfant d'âge préscolaire , DEAD-box RNA helicases/génétique , Variations de nombre de copies de segment d'ADN/génétique , Femelle , Analyse de profil d'expression de gènes , Séquençage nucléotidique à haut débit , Humains , Nourrisson , Facteurs de transcription Krüppel-like/génétique , Mâle , Médulloblastome/traitement médicamenteux , Souris , Souris de lignée NOD , Souris SCID , Données de séquences moléculaires , Protéine du proto-oncogène N-Myc , Transplantation tumorale , Protéines nucléaires/génétique , Protéines oncogènes/génétique , Récepteurs patched , Récepteur Patched-1 , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Régions promotrices (génétique)/génétique , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Pyridines/usage thérapeutique , Récepteurs de surface cellulaire/génétique , Protéines de répression/génétique , Transduction du signal/génétique , Récepteur Smoothened , Telomerase/génétique , Protéine p53 suppresseur de tumeur/génétique , Jeune adulte , Protéine à doigts de zinc Gli2
8.
Hum Gene Ther ; 25(7): 631-41, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-24635742

RÉSUMÉ

Standard integration-proficient lentiviral vectors (IPLVs) are effective at much lower doses than other vector systems and have shown promise for gene therapy of Parkinson's disease (PD). Their main drawback is the risk of insertional mutagenesis. The novel biosafety-enhanced integration-deficient lentiviral vectors (IDLVs) may offer a significant enhancement in biosafety, but have not been previously tested in a model of a major disease. We have assessed biosafety and transduction efficiency of IDLVs in a rat model of PD, using IPLVs as a reference. Genomic insertion of lentivectors injected into the lesioned striatum was studied by linear amplification-mediated polymerase chain reaction (PCR), followed by deep sequencing and insertion site analysis, demonstrating lack of significant IDLV integration. Reporter gene expression studies showed efficient, long-lived, and transcriptionally targeted expression from IDLVs injected ahead of lesioning in the rat striatum, although at somewhat lower expression levels than from IPLVs. Transgenic human glial cell line-derived neurotrophic factor (hGDNF) expression from IDLVs was used for a long-term investigation of lentivector-mediated, transcriptionally targeted neuroprotection in this PD rat model. Vectors were injected before striatal lesioning, and the results showed improvements in nigral dopaminergic neuron survival and behavioral tests regardless of lentiviral integration proficiency, although they confirmed lower expression levels of hGDNF from IDLVs. These data demonstrate the effectiveness of IDLVs in a model of a major disease and indicate that these vectors could provide long-term PD treatment at low dose, combining efficacy and biosafety for targeted central nervous system applications.


Sujet(s)
Neurones dopaminergiques/métabolisme , Thérapie génétique/méthodes , Vecteurs génétiques , Facteur neurotrophique dérivé des cellules gliales , Lentivirus , Neuroprotecteurs/métabolisme , Maladie de Parkinson/thérapie , Animaux , Survie cellulaire/génétique , Modèles animaux de maladie humaine , Neurones dopaminergiques/anatomopathologie , Vecteurs génétiques/génétique , Vecteurs génétiques/pharmacologie , Facteur neurotrophique dérivé des cellules gliales/biosynthèse , Facteur neurotrophique dérivé des cellules gliales/génétique , Humains , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Rats , Rat Sprague-Dawley
9.
Mol Ther ; 22(3): 567-574, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-23941813

RÉSUMÉ

Integration-deficient lentiviral vectors (IDLVs) have been shown to transduce a wide spectrum of target cells and organs in vitro and in vivo and to maintain long-term transgene expression in nondividing cells. However, epigenetic silencing of episomal vector genomes reduces IDLV transgene expression levels and renders these safe vectors less efficient. In this article, we describe for the first time a complete correction of factor IX (FIX) deficiency in hemophilia B mice by IDLVs carrying a novel, highly potent human FIX cDNA. A 50-fold increase in human FIX cDNA potency was achieved by combining two mechanistically independent yet synergistic strategies: (i) optimization of the human FIX cDNA codon usage to increase human FIX protein production per vector genome and (ii) generation of a highly catalytic mutant human FIX protein in which the arginine residue at position 338 was substituted with leucine. The enhanced human FIX activity was not associated with liver damage or with the formation of human FIX-directed inhibitory antibodies and rendered IDLV-treated FIX-knockout mice resistant to a challenging tail-clipping assay. A novel S1 nuclease-based B1-quantitative polymerase chain reaction assay showed low levels of IDLV integration in mouse liver. Overall, this study demonstrates that IDLVs carrying an improved human FIX cDNA safely and efficiently cure hemophilia B in a mouse model.


Sujet(s)
Facteur IX/génétique , Vecteurs génétiques/administration et posologie , Hémophilie B/thérapie , Lentivirus/génétique , Animaux , Arginine/métabolisme , Codon , Modèles animaux de maladie humaine , Facteur IX/métabolisme , Thérapie génétique , Vecteurs génétiques/usage thérapeutique , Hémophilie B/anatomopathologie , Cellules HepG2 , Humains , Leucine/métabolisme , Foie/anatomopathologie , Souris , Souris de lignée C57BL , Souris knockout
10.
Nat Genet ; 45(8): 927-32, 2013 Aug.
Article de Anglais | MEDLINE | ID: mdl-23817572

RÉSUMÉ

Pilocytic astrocytoma, the most common childhood brain tumor, is typically associated with mitogen-activated protein kinase (MAPK) pathway alterations. Surgically inaccessible midline tumors are therapeutically challenging, showing sustained tendency for progression and often becoming a chronic disease with substantial morbidities. Here we describe whole-genome sequencing of 96 pilocytic astrocytomas, with matched RNA sequencing (n = 73), conducted by the International Cancer Genome Consortium (ICGC) PedBrain Tumor Project. We identified recurrent activating mutations in FGFR1 and PTPN11 and new NTRK2 fusion genes in non-cerebellar tumors. New BRAF-activating changes were also observed. MAPK pathway alterations affected all tumors analyzed, with no other significant mutations identified, indicating that pilocytic astrocytoma is predominantly a single-pathway disease. Notably, we identified the same FGFR1 mutations in a subset of H3F3A-mutated pediatric glioblastoma with additional alterations in the NF1 gene. Our findings thus identify new potential therapeutic targets in distinct subsets of pilocytic astrocytoma and childhood glioblastoma.


Sujet(s)
Astrocytome/génétique , Tumeurs du cerveau/génétique , Mutation , Récepteur FGFR1/génétique , Récepteur trkB/génétique , Animaux , Astrocytome/métabolisme , Séquence nucléotidique , Tumeurs du cerveau/métabolisme , Lignée cellulaire , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Points de cassure de chromosome , Chromosomes humains de la paire 6 , Chromosomes humains de la paire 9 , Facteurs de croissance fibroblastique/métabolisme , Humains , Système de signalisation des MAP kinases , Souris , Modèles moléculaires , Protéines de fusion oncogènes/composition chimique , Protéines de fusion oncogènes/génétique , Conformation des protéines , Protéines proto-oncogènes B-raf/composition chimique , Protéines proto-oncogènes B-raf/génétique , Récepteur FGFR1/métabolisme , Récepteur trkB/métabolisme
11.
Cancer Cell ; 23(2): 159-70, 2013 Feb 11.
Article de Anglais | MEDLINE | ID: mdl-23410972

RÉSUMÉ

Early-onset prostate cancer (EO-PCA) represents the earliest clinical manifestation of prostate cancer. To compare the genomic alteration landscapes of EO-PCA with "classical" (elderly-onset) PCA, we performed deep sequencing-based genomics analyses in 11 tumors diagnosed at young age, and pursued comparative assessments with seven elderly-onset PCA genomes. Remarkable age-related differences in structural rearrangement (SR) formation became evident, suggesting distinct disease pathomechanisms. Whereas EO-PCAs harbored a prevalence of balanced SRs, with a specific abundance of androgen-regulated ETS gene fusions including TMPRSS2:ERG, elderly-onset PCAs displayed primarily non-androgen-associated SRs. Data from a validation cohort of > 10,000 patients showed age-dependent androgen receptor levels and a prevalence of SRs affecting androgen-regulated genes, further substantiating the activity of a characteristic "androgen-type" pathomechanism in EO-PCA.


Sujet(s)
Réarrangement des gènes , Génomique , Protéines de fusion oncogènes/génétique , Tumeurs de la prostate/génétique , Récepteurs aux androgènes/génétique , Serine endopeptidases/génétique , Transactivateurs/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Biologie informatique , Séquençage nucléotidique à haut débit , Humains , Mâle , Adulte d'âge moyen , Régulateur transcriptionnel ERG
12.
Nat Methods ; 10(2): 155-61, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23314173

RÉSUMÉ

Transposons and γ-retroviruses have been efficiently used as insertional mutagens in different tissues to identify molecular culprits of cancer. However, these systems are characterized by recurring integrations that accumulate in tumor cells and that hamper the identification of early cancer-driving events among bystander and progression-related events. We developed an insertional mutagenesis platform based on lentiviral vectors (LVVs) by which we could efficiently induce hepatocellular carcinoma (HCC) in three different mouse models. By virtue of the LVV's replication-deficient nature and broad genome-wide integration pattern, LVV-based insertional mutagenesis allowed identification of four previously unknown liver cancer-associated genes from a limited number of integrations. We validated the oncogenic potential of all the identified genes in vivo, with different levels of penetrance. The newly identified genes are likely to play a role in human cancer because they are upregulated, amplified and/or deleted in human HCCs and can predict clinical outcomes of patients.


Sujet(s)
Carcinome hépatocellulaire/génétique , Lentivirus/génétique , Tumeurs du foie/génétique , Mutagenèse par insertion , Oncogènes , Animaux , Inhibiteur p16 de kinase cycline-dépendante/déficit , Vecteurs génétiques , Humains , Souris , Phosphohydrolase PTEN/déficit , Préalbumine/génétique , Récepteur à l'interféron alpha-bêta/déficit
13.
Exp Hematol ; 41(1): 28-38.e3, 2013 Jan.
Article de Anglais | MEDLINE | ID: mdl-22989760

RÉSUMÉ

Retroviral gene marking has been used successfully in preclinical and clinical transplantation settings. Highly sensitive techniques for vector insertion-site determination, such as linear amplification-mediated polymerase chain reaction (LAM-PCR) in conjunction with next-generation sequencing, have been introduced to assess the composition of gene-marked hematopoiesis at a single-cell level. Here we used these novel techniques for directly comparing clonal reconstitution kinetics in mice transplanted with bone-marrow-derived stem cells genetically marked with either a standard, spleen focus-forming virus long terminal repeat (LTR)-driven γ-retroviral, or a lentiviral self-inactivating vector containing an identical but internal spleen focus-forming virus-derived enhancer/promoter. We observed that the use of the lentiviral self-inactivating vector for gene marking was associated with a broader repertoire of differently marked hematopoietic clones. More importantly, we found a significantly higher probability of insertions in growth-promoting, clonal-dominance-associated genes in the spleen focus-forming virus LTR-driven γ-retroviral vector at later time points of analysis. Based on our data, we suggest that the combined use of LAM-PCR and next-generation sequencing represents a potent tool for the analysis of clonal reconstitution kinetics in the context of gene marking with integrated vectors. At the same time, our findings prove that the use of multiple restriction enzymes for LAM-PCR is indispensable to detect most or ideally all individual stem cell clones contributing to hematopoiesis. We have also found that techniques such as quantitative PCR can be helpful to retrospectively analyze reconstitution kinetics for individual hematopoietic stem cell clones. Finally, our results confirm the notion that marking with lentiviral self-inactivating vectors is associated with a lower risk of genotoxicity as compared with γ-retroviral LTR vectors.


Sujet(s)
Gammaretrovirus/génétique , Hématopoïèse , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/virologie , Lentivirus/génétique , Séquences répétées terminales/génétique , Animaux , Clones cellulaires/cytologie , Clones cellulaires/métabolisme , Vecteurs génétiques/génétique , Cellules souches hématopoïétiques/métabolisme , Cinétique , Souris , Réaction de polymérisation en chaîne , Transduction génétique
14.
Mol Ther ; 21(1): 175-84, 2013 Jan.
Article de Anglais | MEDLINE | ID: mdl-22371846

RÉSUMÉ

Gene therapy with ex vivo-transduced hematopoietic stem/progenitor cells may represent a valid therapeutic option for monogenic immunohematological disorders such as Wiskott-Aldrich syndrome (WAS), a primary immunodeficiency associated with thrombocytopenia. We evaluated the preclinical safety and efficacy of human CD34(+) cells transduced with lentiviral vectors (LV) encoding WAS protein (WASp). We first set up and validated a transduction protocol for CD34(+) cells derived from bone marrow (BM) or mobilized peripheral blood (MPB) using a clinical grade, highly purified LV. Robust transduction of progenitor cells was obtained in normal donors and WAS patients' cells, without evidence of toxicity. To study biodistribution of human cells and exclude vector release in vivo, LV-transduced CD34(+) cells were transplanted in immunodeficient mice, showing a normal engraftment and differentiation ability towards transduced lymphoid and myeloid cells in hematopoietic tissues. Vector mobilization to host cells and transmission to germline cells of the LV were excluded by different molecular assays. Analysis of vector integrations showed polyclonal integration patterns in vitro and in human engrafted cells in vivo. In summary, this work establishes the preclinical safety and efficacy of human CD34(+) cells gene therapy for the treatment of WAS.


Sujet(s)
Antigènes CD34/immunologie , Cellules de la moelle osseuse/cytologie , Transplantation de moelle osseuse , Vecteurs génétiques , Lentivirus/génétique , Transduction génétique , Syndrome de Wiskott-Aldrich/thérapie , Animaux , Cellules de la moelle osseuse/immunologie , Souris , Souris knockout
15.
Nature ; 488(7409): 100-5, 2012 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-22832583

RÉSUMÉ

Medulloblastoma is an aggressively growing tumour, arising in the cerebellum or medulla/brain stem. It is the most common malignant brain tumour in children, and shows tremendous biological and clinical heterogeneity. Despite recent treatment advances, approximately 40% of children experience tumour recurrence, and 30% will die from their disease. Those who survive often have a significantly reduced quality of life. Four tumour subgroups with distinct clinical, biological and genetic profiles are currently identified. WNT tumours, showing activated wingless pathway signalling, carry a favourable prognosis under current treatment regimens. SHH tumours show hedgehog pathway activation, and have an intermediate prognosis. Group 3 and 4 tumours are molecularly less well characterized, and also present the greatest clinical challenges. The full repertoire of genetic events driving this distinction, however, remains unclear. Here we describe an integrative deep-sequencing analysis of 125 tumour-normal pairs, conducted as part of the International Cancer Genome Consortium (ICGC) PedBrain Tumor Project. Tetraploidy was identified as a frequent early event in Group 3 and 4 tumours, and a positive correlation between patient age and mutation rate was observed. Several recurrent mutations were identified, both in known medulloblastoma-related genes (CTNNB1, PTCH1, MLL2, SMARCA4) and in genes not previously linked to this tumour (DDX3X, CTDNEP1, KDM6A, TBR1), often in subgroup-specific patterns. RNA sequencing confirmed these alterations, and revealed the expression of what are, to our knowledge, the first medulloblastoma fusion genes identified. Chromatin modifiers were frequently altered across all subgroups. These findings enhance our understanding of the genomic complexity and heterogeneity underlying medulloblastoma, and provide several potential targets for new therapeutics, especially for Group 3 and 4 patients.


Sujet(s)
Tumeurs du cervelet/génétique , Génome humain/génétique , Médulloblastome/génétique , Vieillissement/génétique , Séquence d'acides aminés , Transformation cellulaire néoplasique , Tumeurs du cervelet/classification , Tumeurs du cervelet/diagnostic , Tumeurs du cervelet/anatomopathologie , Enfant , Chromatine/métabolisme , Chromosomes humains/génétique , DEAD-box RNA helicases/génétique , Helicase/génétique , Protéines de liaison à l'ADN/génétique , Génomique , Protéines Hedgehog/métabolisme , Séquençage nucléotidique à haut débit , Histone Demethylases/génétique , Humains , Médulloblastome/classification , Médulloblastome/diagnostic , Médulloblastome/anatomopathologie , Méthylation , Mutation/génétique , Taux de mutation , Protéines tumorales/génétique , Protéines nucléaires/génétique , Protéines de fusion oncogènes/génétique , Récepteurs patched , Récepteur Patched-1 , Phosphoprotein Phosphatases/génétique , Polyploïdie , Récepteurs de surface cellulaire/génétique , Analyse de séquence d'ARN , Transduction du signal , Protéines à domaine boîte-T/génétique , Facteurs de transcription/génétique , Protéines de type Wingless/métabolisme , bêta-Caténine/génétique
16.
Hum Gene Ther Methods ; 23(2): 111-8, 2012 Apr.
Article de Anglais | MEDLINE | ID: mdl-22559057

RÉSUMÉ

Clonality analysis of viral vector-transduced cell populations represents a convincing approach to dissect the physiology of tissue and organ regeneration, to monitor the fate of individual gene-corrected cells in vivo, and to assess vector biosafety. With the decoding of mammalian genomes and the introduction of next-generation sequencing technologies, the demand for automated bioinformatic analysis tools that can rapidly process and annotate vector integration sites is rising. Here, we provide a publicly accessible, graphical user interface-guided automated bioinformatic high-throughput integration site analysis pipeline. Its performance and key features are illustrated on pyrosequenced linear amplification-mediated PCR products derived from one patient previously enrolled in the first lentiviral vector clinical gene therapy study. Analysis includes trimming of vector genome junctions, alignment of genomic sequence fragments to the host genome for the identification of integration sites, and the annotation of nearby genomic elements. Most importantly, clinically relevant features comprise the determination of identical integration sites with respect to different time points or cell lineages, as well as the retrieval of the most prominent cell clones and common integration sites. The resulting output is summarized in tables within a convenient spreadsheet and can be further processed by researchers without profound bioinformatic knowledge.


Sujet(s)
Biologie informatique/méthodes , Thérapie génétique/méthodes , Vecteurs génétiques/génétique , Séquençage nucléotidique à haut débit/méthodes , Lentivirus/génétique , Logiciel , Intégration virale/génétique , Animaux , Séquençage nucléotidique à haut débit/tendances , Humains , Souris , Rats
17.
Methods Mol Biol ; 859: 255-65, 2012.
Article de Anglais | MEDLINE | ID: mdl-22367877

RÉSUMÉ

In gene therapy, viral or nonviral integrating vectors are used to deliver a corrected gene to replace the corresponding defective cellular gene. As vector delivery is (yet) commonly not targeted to a specific site in the host genome, and vector integration may lead to unwanted cellular gene deregulation, the comprehensive analysis of vector locations is a crucial approach to assess vector biosafety and to follow the fate of the gene corrected cells in vivo. The retrieved vector integration sites are unique for each transduced cell clone, thereby serving as a molecular marker and allowing to track distinct cell clones in various samples. Today, several PCR-based methods are available for the identification and characterization of unknown flanking DNA sequences (Mueller and Wold Science 246:780-786, 1989; Paruzynski et al. Nat Protoc 5:1379-1395, 2010; Schmidt et al. Nat Methods 4:1051-1057, 2007; Silver and Keerikatte J Virol 63:1924-1928, 1989). Thereof, the linear amplification-mediated PCR (LAM-PCR) proved to exhibit the highest sensitivity, allowing the detection of miscellaneous vector integration sites in one sample. The broad application spectrum and robustness of LAM-PCR has been approved by its application as a tool for the molecular follow up of gene-modified cells in preclinical and clinical gene therapy trials (Li et al. Science 296:497, 2002; Cartier et al. Science 326:818-823, 2009; Ott et al. Nat Med 12:401-409, 2006; Deichmann et al. J Clin Invest 117:2225-2232, 2007). The combination of LAM-PCR and next-generation sequencing (NGS) platforms offers the opportunity to study the clonal inventory and pharmacokinetics in clinical gene therapy studies.


Sujet(s)
Thérapie génétique/méthodes , Séquençage nucléotidique à haut débit/méthodes , Mutagenèse par insertion , Réaction de polymérisation en chaîne/méthodes , Analyse de séquence d'ADN/méthodes , Chromatographie sur gel , Clonage moléculaire , ADN/génétique , ADN/isolement et purification , Clivage de l'ADN , Humains
18.
Methods Enzymol ; 507: 187-98, 2012.
Article de Anglais | MEDLINE | ID: mdl-22365775

RÉSUMÉ

X-linked adrenoleukodystrophy (X-ALD) is a severe genetic demyelinating disease caused by a deficiency in ALD protein, an adenosine triphosphate-binding cassette transporter encoded by the ABCD1 gene. When performed at an early stage of the disease, allogeneic hematopoietic stem cell transplantation (HCT) can arrest the progression of cerebral demyelinating lesions. To overcome the limitations of allogeneic HCT, hematopoietic stem cell (HSC) gene therapy strategy aiming to perform autologous transplantation of lentivirally corrected cells was developed. We demonstrated the preclinical feasibility of HSC gene therapy for ALD based on the correction of CD34+ cells from X-ALD patients using an HIV1-derived lentiviral vector. These results prompted us to initiate an HSC gene therapy trial in two X-ALD patients who had developed progressive cerebral demyelination, were candidates for allogeneic HCT, but had no HLA-matched donors or cord blood. Autologous CD34+ cells were purified from the peripheral blood after G-CSF stimulation, genetically corrected ex vivo with a lentiviral vector encoding wild-type ABCD1 cDNA, and then reinfused into the patients after they had received full myeloablative conditioning. Over 3 years of follow-up, the hematopoiesis remained polyclonal in the two patients treated with 7-14% of granulocytes, monocytes, and T and B lymphocytes expressing the lentivirally encoded ALD protein. There was no evidence of clonal dominance or skewing based on the retrieval of lentiviral insertion repertoire in different hematopoietic lineages by deep sequencing. Cerebral demyelination was arrested 14 and 16months, respectively, in the two treated patients, without further progression up to the last follow-up, a clinical outcome that is comparable to that observed after allogeneic HCT. Longer follow-up of these two treated patients and HSC gene therapy performed in additional ALD patients are however needed to evaluate the safety and efficacy of lentiviral HSC gene therapy in cerebral forms of X-ALD.


Sujet(s)
Adrénoleucodystrophie/thérapie , Thérapie génétique , Lentivirus/génétique , Membre-1 de la sous-famille D de transporteurs à cassette liant l'ATP , Transporteurs ABC/génétique , Enfant , Techniques de transfert de gènes , Vecteurs génétiques , Cellules HEK293 , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques/virologie , Humains , Mâle , Résultat thérapeutique
19.
PLoS One ; 6(10): e24247, 2011.
Article de Anglais | MEDLINE | ID: mdl-22022353

RÉSUMÉ

Vectors based on γ-retroviruses or lentiviruses have been shown to stably express therapeutical transgenes and effectively cure different hematological diseases. Molecular follow up of the insertional repertoire of gene corrected cells in patients and preclinical animal models revealed different integration preferences in the host genome including clusters of integrations in small genomic areas (CIS; common integrations sites). In the majority, these CIS were found in or near genes, with the potential to influence the clonal fate of the affected cell. To determine whether the observed degree of clustering is statistically compatible with an assumed standard model of spatial distribution of integrants, we have developed various methods and computer programs for γ-retroviral and lentiviral integration site distribution. In particular, we have devised and implemented mathematical and statistical approaches for comparing two experimental samples with different numbers of integration sites with respect to the propensity to form CIS as well as for the analysis of coincidences of integration sites obtained from different blood compartments. The programs and statistical tools described here are available as workspaces in R code and allow the fast detection of excessive clustering of integration sites from any retrovirally transduced sample and thus contribute to the assessment of potential treatment-related risks in preclinical and clinical retroviral gene therapy studies.


Sujet(s)
Vecteurs génétiques/génétique , Mutagenèse par insertion/méthodes , Retroviridae/génétique , Logiciel , Simulation numérique , Bases de données génétiques , Humains , Modèles génétiques
20.
Mol Ther ; 19(11): 2092-101, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21878903

RÉSUMÉ

X-linked chronic granulomatous disease (CGD) is an inherited immunodeficiency caused by a defect in the gp91(phox) gene. In an effort to treat X-CGD, we investigated the safety and efficacy of gene therapy using a retroviral vector, MT-gp91. Two X-CGD patients received autologous CD34(+) cells transduced with MT-gp91 after a conditioning regimen consisting of fludarabine and busulfan. The level of gene-marked cells was highest at day 21 (8.3 and 11.7% in peripheral blood cells) but decreased to 0.08 and 0.5%, respectively, 3 years after gene transfer. The level of functionally corrected cells, as determined by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase assay, reached a peak at day 17 (6.5% patient 1 (P1) and 14.3% patient 2 (P2) of total granulocytes) and declined to 0.05% (P1) and 0.21% (P2), 3 years later. Some retroviral vectors were found to have integrated within or close to the proto-oncogenes MDS1-EVI1, PRDM16, and CCND2; however, no abnormal cell expansion or related hematological malignancy was observed. Overall, the gene transfer procedure did not produce any serious adverse effects and was able to convert a significant fraction of blood cells to biologically functional cells, albeit for a short period of time.


Sujet(s)
Thérapie génétique , Vecteurs génétiques , Granulomatose septique chronique/thérapie , Retroviridae/génétique , Adolescent , Enfant , Analyse de profil d'expression de gènes , Vecteurs génétiques/effets indésirables , Transplantation de cellules souches hématopoïétiques/effets indésirables , Cellules souches hématopoïétiques/métabolisme , Humains , Mâle , Glycoprotéines membranaires/génétique , NADPH Oxidase 2 , NADPH oxidase/génétique , Transduction génétique , Résultat thérapeutique , Intégration virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...