Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Br J Clin Pharmacol ; 90(5): 1203-1212, 2024 May.
Article in English | MEDLINE | ID: mdl-38565322

ABSTRACT

Great advances have been made in the knowledge of development and regulatory approval of medicinal product containing genetically modified cells. Although a guideline has been available in the EU since 2012, the current updated version provides a useful guide to developers and professionals involved in the regulatory process of these medicines. This article presents the main issues communicated in that guidance, the regulators' insights and a commentary from the academic developers' point of view.


Subject(s)
Drug Approval , European Union , Guidelines as Topic , Humans , Drug Approval/legislation & jurisprudence , Animals
2.
Lancet ; 399(10322): 372-383, 2022 01 22.
Article in English | MEDLINE | ID: mdl-35065785

ABSTRACT

BACKGROUND: Effective treatment for metachromatic leukodystrophy (MLD) remains a substantial unmet medical need. In this study we investigated the safety and efficacy of atidarsagene autotemcel (arsa-cel) in patients with MLD. METHODS: This study is an integrated analysis of results from a prospective, non-randomised, phase 1/2 clinical study and expanded-access frameworks. 29 paediatric patients with pre-symptomatic or early-symptomatic early-onset MLD with biochemical and molecular confirmation of diagnosis were treated with arsa-cel, a gene therapy containing an autologous haematopoietic stem and progenitor cell (HSPC) population transduced ex vivo with a lentiviral vector encoding human arylsulfatase A (ARSA) cDNA, and compared with an untreated natural history (NHx) cohort of 31 patients with early-onset MLD, matched by age and disease subtype. Patients were treated and followed up at Ospedale San Raffaele, Milan, Italy. The coprimary efficacy endpoints were an improvement of more than 10% in total gross motor function measure score at 2 years after treatment in treated patients compared with controls, and change from baseline of total peripheral blood mononuclear cell (PBMC) ARSA activity at 2 years after treatment compared with values before treatment. This phase 1/2 study is registered with ClinicalTrials.gov, NCT01560182. FINDINGS: At the time of analyses, 26 patients treated with arsa-cel were alive with median follow-up of 3·16 years (range 0·64-7·51). Two patients died due to disease progression and one due to a sudden event deemed unlikely to be related to treatment. After busulfan conditioning, all arsa-cel treated patients showed sustained multilineage engraftment of genetically modified HSPCs. ARSA activity in PBMCs was significantly increased above baseline 2 years after treatment by a mean 18·7-fold (95% CI 8·3-42·2; p<0·0001) in patients with the late-infantile variant and 5·7-fold (2·6-12·4; p<0·0001) in patients with the early-juvenile variant. Mean differences in total scores for gross motor function measure between treated patients and age-matched and disease subtype-matched NHx patients 2 years after treatment were significant for both patients with late-infantile MLD (66% [95% CI 48·9-82·3]) and early-juvenile MLD (42% [12·3-71·8]). Most treated patients progressively acquired motor skills within the predicted range of healthy children or had stabilised motor performance (maintaining the ability to walk). Further, most displayed normal cognitive development and prevention or delay of central and peripheral demyelination and brain atrophy throughout follow-up; treatment benefits were particularly apparent in patients treated before symptom onset. The infusion was well tolerated and there was no evidence of abnormal clonal proliferation or replication-competent lentivirus. All patients had at least one grade 3 or higher adverse event; most were related to conditioning or to background disease. The only adverse event related to arsa-cel was the transient development of anti-ARSA antibodies in four patients, which did not affect clinical outcomes. INTERPRETATION: Treatment with arsa-cel resulted in sustained, clinically relevant benefits in children with early-onset MLD by preserving cognitive function and motor development in most patients, and slowing demyelination and brain atrophy. FUNDING: Orchard Therapeutics, Fondazione Telethon, and GlaxoSmithKline.


Subject(s)
Cerebroside-Sulfatase/genetics , Hematopoietic Stem Cell Transplantation , Lentivirus/genetics , Leukodystrophy, Metachromatic , Age of Onset , Child , Child, Preschool , Female , Genetic Therapy , Genetic Vectors , Humans , Italy , Leukodystrophy, Metachromatic/genetics , Leukodystrophy, Metachromatic/therapy , Male , Prospective Studies , Treatment Outcome
3.
J Clin Immunol ; 42(8): 1742-1747, 2022 11.
Article in English | MEDLINE | ID: mdl-35945378

ABSTRACT

X-linked chronic granulomatous disease is a rare disease caused by mutations in the CYBB gene. While more extensive knowledge is available on genetics, pathogenesis, and possible therapeutic options, mitochondrial activity and its implications on patient monitoring are still not well-characterized. We have developed a novel protocol to study mitochondrial activity on whole blood of XCGD patients before and after transplantation, as well as on XCGD carriers. Here we present results of these analyses and of the restoration of mitochondrial activity in hyperinflamed X-linked Chronic Granulomatous Disease after hematopoietic stem cell transplantation. Moreover, we show a strong direct correlation between mitochondrial activity, chimerism, and DHR monitored before and after transplantation and in XCGD carriers. In conclusion, based on these findings, we suggest testing this new ready-to-use marker to better characterize patients before and after treatment and to investigate disease expression in carriers.


Subject(s)
Granulomatous Disease, Chronic , Hematopoietic Stem Cell Transplantation , Humans , Granulomatous Disease, Chronic/diagnosis , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/therapy , Chimerism , Phagocytes , Heterozygote
4.
Mol Ther ; 29(1): 86-102, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33010230

ABSTRACT

Chronic granulomatous disease (CGD) is a rare inherited disorder due to loss-of-function mutations in genes encoding the NADPH oxidase subunits. Hematopoietic stem and progenitor cell (HSPC) gene therapy (GT) using regulated lentiviral vectors (LVs) has emerged as a promising therapeutic option for CGD patients. We performed non-clinical Good Laboratory Practice (GLP) and laboratory-grade studies to assess the safety and genotoxicity of LV targeting myeloid-specific Gp91phox expression in X-linked chronic granulomatous disease (XCGD) mice. We found persistence of gene-corrected cells for up to 1 year, restoration of Gp91phox expression and NADPH oxidase activity in XCGD phagocytes, and reduced tissue inflammation after LV-mediated HSPC GT. Although most of the mice showed no hematological or biochemical toxicity, a small subset of XCGD GT mice developed T cell lymphoblastic lymphoma (2.94%) and myeloid leukemia (5.88%). No hematological malignancies were identified in C57BL/6 mice transplanted with transduced XCGD HSPCs. Integration pattern analysis revealed an oligoclonal composition with rare dominant clones harboring vector insertions near oncogenes in mice with tumors. Collectively, our data support the long-term efficacy of LV-mediated HSPC GT in XCGD mice and provide a safety warning because the chronic inflammatory XCGD background may contribute to oncogenesis.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/therapy , Hematologic Neoplasms/etiology , Lentivirus/genetics , Animals , Disease Models, Animal , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Granulomatous Disease, Chronic/genetics , Humans , Mice , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Time Factors , Treatment Outcome
5.
Mol Ther ; 24(10): 1873-1880, 2016 10.
Article in English | MEDLINE | ID: mdl-27456061

ABSTRACT

Chronic granulomatous disease (CGD) is a primary immunodeficiency due to a deficiency in one of the subunits of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. CGD patients are characterized by an increased susceptibility to bacterial and fungal infections, and to granuloma formation due to the excessive inflammatory responses. Several gene therapy approaches with lentiviral vectors have been proposed but there is a lack of in vivo data on the ability to control infections and inflammation. We set up a mouse model of acute infection that closely mimic the airway infection in CGD patients. It involved an intratracheal injection of a methicillin-sensitive reference strain of S. aureus. Gene therapy, with hematopoietic stem cells transduced with regulated lentiviral vectors, restored the functional activity of NADPH oxidase complex (with 20-98% of dihydrorhodamine positive granulocytes and monocytes) and saved mice from death caused by S. aureus, significantly reducing the bacterial load and lung damage, similarly to WT mice even at low vector copy number. When challenged, gene therapy-treated XCGD mice showed correction of proinflammatory cytokines and chemokine imbalance at levels that were comparable to WT. Examined together, our results support the clinical development of gene therapy protocols using lentiviral vectors for the protection against infections and inflammation.


Subject(s)
Genetic Therapy/methods , Granulomatous Disease, Chronic/complications , Hematopoietic Stem Cell Transplantation/methods , Membrane Glycoproteins/genetics , NADPH Oxidases/genetics , Pneumonia, Staphylococcal/therapy , Staphylococcus aureus/physiology , Animals , Bacterial Load , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Genetic Vectors/administration & dosage , Granulomatous Disease, Chronic/genetics , Hematopoietic Stem Cells/virology , Humans , Lentivirus/genetics , Mice , NADPH Oxidase 2 , Pneumonia, Staphylococcal/genetics , Pneumonia, Staphylococcal/microbiology
6.
Mol Ther ; 22(8): 1472-1483, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24869932

ABSTRACT

Regulated transgene expression may improve the safety and efficacy of hematopoietic stem cell (HSC) gene therapy. Clinical trials for X-linked chronic granulomatous disease (X-CGD) employing gammaretroviral vectors were limited by insertional oncogenesis or lack of persistent engraftment. Our novel strategy, based on regulated lentiviral vectors (LV), targets gp91(phox) expression to the differentiated myeloid compartment while sparing HSC, to reduce the risk of genotoxicity and potential perturbation of reactive oxygen species levels. Targeting was obtained by a myeloid-specific promoter (MSP) and posttranscriptional, microRNA-mediated regulation. We optimized both components in human bone marrow (BM) HSC and their differentiated progeny in vitro and in a xenotransplantation model, and generated therapeutic gp91(phox) expressing LVs for CGD gene therapy. All vectors restored gp91(phox) expression and function in human X-CGD myeloid cell lines, primary monocytes, and differentiated myeloid cells. While unregulated LVs ectopically expressed gp91(phox) in CD34(+) cells, transcriptionally and posttranscriptionally regulated LVs substantially reduced this off-target expression. X-CGD mice transplanted with transduced HSC restored gp91(phox) expression, and MSP-driven vectors maintained regulation during BM development. Combining transcriptional (SP146.gp91-driven) and posttranscriptional (miR-126-restricted) targeting, we achieved high levels of myeloid-specific transgene expression, entirely sparing the CD34(+) HSC compartment. This dual-targeted LV construct represents a promising candidate for further clinical development.


Subject(s)
Genetic Therapy/methods , Granulomatous Disease, Chronic/therapy , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/virology , Membrane Glycoproteins/metabolism , MicroRNAs/genetics , NADPH Oxidases/metabolism , Animals , Antigens, CD34/metabolism , Cell Line , Cells, Cultured , Combined Modality Therapy , Disease Models, Animal , Genetic Vectors/therapeutic use , Granulomatous Disease, Chronic/pathology , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/genetics , Mice , Myeloid Cells/metabolism , NADPH Oxidase 2
7.
J Inherit Metab Dis ; 37(4): 525-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24619149

ABSTRACT

In the last years important progress has been made in the treatment of several primary immunodeficiency disorders (PIDs) with gene therapy. Hematopoietic stem cell (HSC) gene therapy indeed represents a valid alternative to conventional transplantation when a compatible donor is not available and recent success confirmed the great potential of this approach. First clinical trials performed with gamma retroviral vectors were promising and guaranteed clinical benefits to the patients. On the other hand, the outcome of severe adverse events as the development of hematological abnormalities highlighted the necessity to develop a safer platform to deliver the therapeutic gene. Self-inactivating (SIN) lentiviral vectors (LVVs) were studied to overcome this hurdle through their preferable integration pattern into the host genome. In this review, we describe the recent advancements achieved both in vitro and at preclinical level with LVVs for the treatment of Wiskott-Aldrich syndrome (WAS), chronic granulomatous disease (CGD), ADA deficiency (ADA-SCID), Artemis deficiency, RAG1/2 deficiency, X-linked severe combined immunodeficiency (γchain deficiency, SCIDX1), X-linked lymphoproliferative disease (XLP) and immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/therapeutic use , Immunologic Deficiency Syndromes/therapy , Lentivirus/genetics , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Agammaglobulinemia/genetics , Agammaglobulinemia/therapy , Animals , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/therapy , Humans , Immunologic Deficiency Syndromes/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/therapy
10.
J Leukoc Biol ; 100(5): 1071-1079, 2016 11.
Article in English | MEDLINE | ID: mdl-27256569

ABSTRACT

H2O2 acts as a second messenger in key signaling circuits, transiently modulating tyrosine phosphatases and kinases. We investigated its origin, membrane transport, and functional role during B cell activation and differentiation. Our data identified NADPH-oxidase 2 as the main source of H2O2 and aquaporin 8 as a transport facilitator across the plasma membrane. On aquaporin 8 silencing, inducible B lymphoma cells responded poorly to TLR and BCR stimulation. Their differentiation was severely impaired, as demonstrated by retarded onset of IgM polymerization, low amounts of IgM secretion, and prolonged BCR expression on the cell surface. A silencing-resistant aquaporin 8 rescued responsiveness, confirming that the import of H2O2 across the membrane is essential for B cell activation. The addition of exogenous catalase to primary B splenocytes severely impaired the tyrosine phosphorylation induced by BCR cross-linking, as did the absence of NOX2 in a murine model of chronic granulomatous disease. Importantly, re-expression of gp91phox through gene therapy restored the specific B cell signaling deficiency in NOX2-/- cells. Thus, efficient induction of B cell activation and differentiation requires intact H2O2 fluxes across the plasma membrane for signal amplification.


Subject(s)
Aquaporins/physiology , B-Lymphocytes/metabolism , Hydrogen Peroxide/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Animals , Aquaporins/antagonists & inhibitors , Aquaporins/genetics , Biological Transport , Bone Marrow Transplantation , Catalase/pharmacology , Cell Differentiation , Cell Line, Tumor , Cell Membrane/metabolism , Disease Models, Animal , Granulomatous Disease, Chronic , Lymphocyte Activation , Lymphoma, B-Cell/pathology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Phosphorylation/drug effects , Plasma Cells/pathology , Protein Processing, Post-Translational/drug effects , RNA Interference , Receptors, Antigen, B-Cell/immunology , Recombinant Fusion Proteins/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL