Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 613
Filtrar
1.
Inn Med (Heidelb) ; 65(6): 617-623, 2024 Jun.
Artículo en Alemán | MEDLINE | ID: mdl-38748280

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is a severe monogenic hereditary disease with early manifestation and a progressive course. Treatment options have so far been limited. Gene therapy opens up new options for DMD patients. OBJECTIVES: Against the background of a further death following DMD gene therapy, the side effects and risks of the gene therapeutics already approved or undergoing clinical trials will be evaluated and alternative gene therapeutics will be described. Based thereon, the future of DMD gene therapy will be discussed. CURRENT DATA: For the first time, in June 2023, delandistrogene moxeparvovec (SRP-9001), a gene replacement therapy based on an adeno-associated virus (AAV) vector, was approved in the USA for children aged 4-5 years with DMD. Other promising gene therapies are in preclinical development or clinical trials, including CRISPR/Cas9-mediated strategies to restore dystrophin expression. Two deaths following DMD gene therapy with high-dose AAV vectors were attributed to AAV-mediated immune responses. The pre-existing disease underlying the therapy is most likely involved in the fatal AAV toxicity. CONCLUSIONS: Although gene therapy applications of AAV vectors are generally considered safe, the systemic administration of high vector doses can lead to severe side effects with a potentially fatal outcome in individual patients, especially after activation of the immune system. In the future, new methods for immunosuppression, reduction of AAV dose and alternative vectors will therefore increasingly come to the fore.


Asunto(s)
Dependovirus , Terapia Genética , Vectores Genéticos , Distrofia Muscular de Duchenne , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Humanos , Terapia Genética/efectos adversos , Terapia Genética/métodos , Dependovirus/genética , Vectores Genéticos/genética , Vectores Genéticos/efectos adversos , Preescolar , Niño , Masculino
2.
Hum Gene Ther ; 35(13-14): 425-438, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38251650

RESUMEN

Adeno-associated virus (AAV) vectors represent a novel tool for the delivery of genetic therapeutics and enable the treatment of a wide range of diseases. Success of this new modality is challenged, however, by cases of immune-related toxicities that complicate the clinical management of patients and potentially limit the therapeutic efficacy of AAV gene therapy. While significant progress has been made to manage immune-related liver enzyme elevations following systemic AAV delivery in humans, recent clinical trials utilizing high vector doses have highlighted a new challenge to AAV gene transfer-activation of the complement system. While current in vitro models implicate AAV-specific antibodies in the initiation of the classical complement pathway, evidence from in vivo pre-clinical and clinical studies suggests that the alternative pathway also contributes to complement activation. A convergence of AAV-specific, environmental, and patient-specific factors shaping complement responses likely contributes to differential outcomes seen in clinical trials, from priming of the adaptive immune system to serious adverse events such as hepatotoxicity and thrombotic microangiopathy. Research focused on the interplay of patient-specific and AAV-related factors driving complement activation is needed to understand and identify critical components in the complement cascade to target and devise strategies to mitigate vector-related immune responses.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Dependovirus , Terapia Genética , Vectores Genéticos , Dependovirus/genética , Dependovirus/inmunología , Humanos , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Terapia Genética/métodos , Terapia Genética/efectos adversos , Proteínas del Sistema Complemento/inmunología , Animales , Técnicas de Transferencia de Gen
3.
Hum Gene Ther ; 35(3-4): 93-103, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38185849

RESUMEN

Clinical trials of adeno-associated virus (AAV)-based gene therapy have made remarkable progress in recent years. We aimed to perform a systematic review and meta-analysis of the literature to assess the efficacy and safety of AAV-based gene therapy for hemophilia. We systematically searched the Web of Science, Embase, PubMed, and the Cochrane Database of Systematic Reviews databases, for clinical trials involving patients diagnosed with hemophilia and treated with AAV-mediated gene therapy. Data on the annualized bleeding rate (ABR), annualized infusion rate (AIR), the incidence of treatment-related adverse events (TRAEs), severe adverse events (SAEs), and alanine aminotransferase (ALT) elevation were extracted as our outcomes. A total of 12 articles from 11 clinical trials were selected from 868 articles for meta-analysis. Pooled analyses showed that AAV-based gene therapy in hemophilia patients reduced the number of bleeding events and the number of factor infusion events by an approximate average of 7 per year and 103 per year, respectively. Eighty percent, 18%, and 63% of hemophilia patients had elevated TRAE, SAE, and ALT levels, respectively. Moreover, subgroup analysis found a significant reduction in ABR and AIR 2-3 years after the therapy. Additional findings that were not pooled including coagulation factor activity are presented in the accompanying tables. Our analysis supported the efficacy and safety of AAV-mediated gene therapy for hemophilia, providing evidence for its application as a therapeutic option for widespread clinical use in hemophilia patients in the future.


Asunto(s)
Hemofilia A , Hemofilia B , Humanos , Dependovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Hemofilia A/genética , Hemofilia A/terapia , Hemofilia B/genética , Hemofilia B/terapia , Hemorragia/genética , Hemorragia/terapia
4.
Med ; 4(12): 855-856, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38070478

RESUMEN

Gene therapies, notably those leveraging CRISPR and adeno-associated virus vectors (AAVs), have risen to the forefront of potential treatments for neuromuscular disorders. The recent demise of a Duchenne muscular dystrophy (DMD) patient following a trial utilizing CRISPR transactivation with AAV has cast a spotlight on the potential risks associated with these approaches.1.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Distrofia Muscular de Duchenne , Humanos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Dependovirus/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Terapia Genética/efectos adversos
5.
Mol Ther ; 31(11): 3123-3126, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37822079

RESUMEN

High-dose systemic gene therapy with adeno-associated virus (AAV) is in clinical trials to treat various inherited diseases. Despite remarkable success in spinal muscular atrophy and promising results in other diseases, fatality has been observed due to liver, kidney, heart, or lung failure. Innate and adaptive immune responses to the vector play a critical role in the toxicity. Host factors also contribute to patient death. This mini-review summarizes clinical findings and calls for concerted efforts from all stakeholders to better understand the mechanisms underlying lethality in AAV gene therapy and to develop effective strategies to prevent/treat high-dose systemic AAV-gene-therapy-induced immunotoxicity.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Humanos , Distrofina/genética , Dependovirus/genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Inmunidad Humoral , Distrofia Muscular de Duchenne/genética , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética
6.
N Engl J Med ; 387(25): 2344-2355, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36546626

RESUMEN

BACKGROUND: The DNA-repair enzyme Artemis is essential for rearrangement of T- and B-cell receptors. Mutations in DCLRE1C, which encodes Artemis, cause Artemis-deficient severe combined immunodeficiency (ART-SCID), which is poorly responsive to allogeneic hematopoietic-cell transplantation. METHODS: We carried out a phase 1-2 clinical study of the transfusion of autologous CD34+ cells, transfected with a lentiviral vector containing DCLRE1C, in 10 infants with newly diagnosed ART-SCID. We followed them for a median of 31.2 months. RESULTS: Marrow harvest, busulfan conditioning, and lentiviral-transduced CD34+ cell infusion produced the expected grade 3 or 4 adverse events. All the procedures met prespecified criteria for feasibility at 42 days after infusion. Gene-marked T cells were detected at 6 to 16 weeks after infusion in all the patients. Five of 6 patients who were followed for at least 24 months had T-cell immune reconstitution at a median of 12 months. The diversity of T-cell receptor ß chains normalized by 6 to 12 months. Four patients who were followed for at least 24 months had sufficient B-cell numbers, IgM concentration, or IgM isohemagglutinin titers to permit discontinuation of IgG infusions. Three of these 4 patients had normal immunization responses, and the fourth has started immunizations. Vector insertion sites showed no evidence of clonal expansion. One patient who presented with cytomegalovirus infection received a second infusion of gene-corrected cells to achieve T-cell immunity sufficient for viral clearance. Autoimmune hemolytic anemia developed in 4 patients 4 to 11 months after infusion; this condition resolved after reconstitution of T-cell immunity. All 10 patients were healthy at the time of this report. CONCLUSIONS: Infusion of lentiviral gene-corrected autologous CD34+ cells, preceded by pharmacologically targeted low-exposure busulfan, in infants with newly diagnosed ART-SCID resulted in genetically corrected and functional T and B cells. (Funded by the California Institute for Regenerative Medicine and the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT03538899.).


Asunto(s)
Terapia Genética , Inmunodeficiencia Combinada Grave , Humanos , Lactante , Busulfano/uso terapéutico , Terapia Genética/efectos adversos , Terapia Genética/métodos , Inmunoglobulina M , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Enzimas Reparadoras del ADN/deficiencia , Enzimas Reparadoras del ADN/genética , Antígenos CD34/administración & dosificación , Antígenos CD34/inmunología , Trasplante Autólogo/efectos adversos , Trasplante Autólogo/métodos , Lentivirus , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Vectores Genéticos/uso terapéutico , Linfocitos T/inmunología , Linfocitos B/inmunología
7.
Front Immunol ; 13: 1001263, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36389770

RESUMEN

Recombinant Adeno-associated virus (rAAV) is one of the main delivery vectors for gene therapy. To assess immunogenicity, toxicity, and features of AAV gene therapy in clinical settings, a meta-analysis of 255 clinical trials was performed. A total of 7,289 patients are planned to be dosed. AAV2 was the most dominantly used serotype (29.8%, n=72), and 8.3% (n=20) of trials used engineered capsids. 38.7% (n=91) of trials employed neutralizing antibody assays for patient enrollment, while 15.3% (n=36) used ELISA-based total antibody assays. However, there was high variability in the eligibility criteria with cut-off tiers ranging from 1:1 to 1:1,600. To address potential immunogenicity, 46.3% (n=118) of trials applied immunosuppressants (prophylactic or reactive), while 32.7% (n=18) of CNS and 37.5% (n=24) of ocular-directed trials employed immunosuppressants, possibly due to the immune-privileged status of CNS and retina. There were a total of 11 patient deaths across 8 trials, and 18 out of 30 clinical holds were due to toxicity findings in clinical studies. 30.6% (n=78) of trials had treatment-emergent serious adverse events (TESAEs), with hepatotoxicity and thrombotic microangiopathy (systemic delivery) and neurotoxicity (CNS delivery) being the most prominent. Additionally, the durability of gene therapy may be impacted by two distinct decline mechanisms: 1) rapid decline presumably due to immune responses; or 2) gradual decline due to vector dilution. The durability varied significantly depending on disease indication, dose, serotypes, and patient individuals. Most CNS (90.0%) and muscle trials (73.3%) achieved durable transgene expression, while only 43.6% of ocular trials had sustained clinical outcomes. The rAAV production system can affect rAAV quality and thus immunogenicity and toxicity. Out of 186 trials that have disclosed production system information, 63.0% (n=126) of trials used the transient transfection of the HEK293/HEK293T system, while 18.0% (n=36) applied the baculovirus/Sf9 (rBac/Sf9) system. There were no significant differences in TESAEs and durability between AAV generated by rBac/Sf9 and HEK293/HEK293T systems. In summary, rAAV immunogenicity and toxicity poses significant challenges for clinical development of rAAV gene therapies, and it warrants collaborative efforts to standardize monitoring/measurement methods, design novel strategies to overcome immune responses, and openly share relevant information.


Asunto(s)
Dependovirus , Vectores Genéticos , Humanos , Dependovirus/genética , Células HEK293 , Vectores Genéticos/efectos adversos , Terapia Genética/efectos adversos , Terapia Genética/métodos , Inmunosupresores/efectos adversos
8.
Front Immunol ; 12: 753467, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777364

RESUMEN

Adeno-associated viruses (AAV) have emerged as the lead vector in clinical trials and form the basis for several approved gene therapies for human diseases, mainly owing to their ability to sustain robust and long-term in vivo transgene expression, their amenability to genetic engineering of cargo and capsid, as well as their moderate toxicity and immunogenicity. Still, recent reports of fatalities in a clinical trial for a neuromuscular disease, although linked to an exceptionally high vector dose, have raised new caution about the safety of recombinant AAVs. Moreover, concerns linger about the presence of pre-existing anti-AAV antibodies in the human population, which precludes a significant percentage of patients from receiving, and benefitting from, AAV gene therapies. These concerns are exacerbated by observations of cellular immune responses and other adverse events, including detrimental off-target transgene expression in dorsal root ganglia. Here, we provide an update on our knowledge of the immunological and molecular race between AAV (the "hedgehog") and its human host (the "hare"), together with a compendium of state-of-the-art technologies which provide an advantage to AAV and which, thus, promise safer and more broadly applicable AAV gene therapies in the future.


Asunto(s)
Anticuerpos Antivirales/inmunología , Dependovirus/inmunología , Terapia Genética , Vectores Genéticos/inmunología , Inmunidad Adaptativa , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/biosíntesis , Cápside/efectos de los fármacos , Cápside/inmunología , Ensayos Clínicos como Asunto , Dependovirus/clasificación , Dependovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Humanos , Tolerancia Inmunológica , Inmunidad Celular , Inmunidad Innata , Memoria Inmunológica , Subgrupos Linfocitarios/inmunología , Especificidad de Órganos , Serotipificación , Transgenes
9.
Int J Mol Sci ; 22(19)2021 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-34639132

RESUMEN

Novel coronavirus SARS-CoV-2 has resulted in a global pandemic with worldwide 6-digit infection rates and thousands of death tolls daily. Enormous efforts are undertaken to achieve high coverage of immunization to reach herd immunity in order to stop the spread of SARS-CoV-2 infection. Several SARS-CoV-2 vaccines based on mRNA, viral vectors, or inactivated SARS-CoV-2 virus have been approved and are being applied worldwide. However, the recent increased numbers of normally very rare types of thromboses associated with thrombocytopenia have been reported, particularly in the context of the adenoviral vector vaccine ChAdOx1 nCoV-19 from Astra Zeneca. The statistical prevalence of these side effects seems to correlate with this particular vaccine type, i.e., adenoviral vector-based vaccines, but the exact molecular mechanisms are still not clear. The present review summarizes current data and hypotheses for molecular and cellular mechanisms into one integrated hypothesis indicating that coagulopathies, including thromboses, thrombocytopenia, and other related side effects, are correlated to an interplay of the two components in the vaccine, i.e., the spike antigen and the adenoviral vector, with the innate and immune systems, which under certain circumstances can imitate the picture of a limited COVID-19 pathological picture.


Asunto(s)
Vacunas contra la COVID-19/efectos adversos , COVID-19/prevención & control , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Trombocitopenia/etiología , Trombosis/etiología , Adenoviridae/inmunología , Animales , COVID-19/inmunología , Vacunas contra la COVID-19/inmunología , ChAdOx1 nCoV-19 , Vectores Genéticos/efectos adversos , Vectores Genéticos/inmunología , Humanos , Púrpura Trombocitopénica Idiopática/etiología , Púrpura Trombocitopénica Idiopática/inmunología , Glicoproteína de la Espiga del Coronavirus/efectos adversos , Trombocitopenia/inmunología , Trombosis/inmunología , Vacunación/efectos adversos
10.
Nat Med ; 27(10): 1701-1711, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34608334

RESUMEN

Spinal muscular atrophy type 1 (SMA1) is a debilitating neurodegenerative disease resulting from survival motor neuron 1 gene (SMN1) deletion/mutation. Onasemnogene abeparvovec (formerly AVXS-101) is a gene therapy that restores SMN production via one-time systemic administration. The present study demonstrates widespread biodistribution of vector genomes and transgenes throughout the central nervous system (CNS) and peripheral organs, after intravenous administration of an AAV9-mediated gene therapy. Two symptomatic infants with SMA1 enrolled in phase III studies received onasemnogene abeparvovec. Both patients died of respiratory complications unrelated to onasemnogene abeparvovec. One patient had improved motor function and the other died shortly after administration before appreciable clinical benefit could be observed. In both patients, onasemnogene abeparvovec DNA and messenger RNA distribution were widespread among peripheral organs and in the CNS. The greatest concentration of vector genomes was detected in the liver, with an increase over that detected in CNS tissues of 300-1,000-fold. SMN protein, which was low in an untreated SMA1 control, was clearly detectable in motor neurons, brain, skeletal muscle and multiple peripheral organs in treated patients. These data support the fact that onasemnogene abeparvovec has effective distribution, transduction and expression throughout the CNS after intravenous administration and restores SMN expression in humans.


Asunto(s)
Productos Biológicos/efectos adversos , Terapia Genética/efectos adversos , Proteínas Recombinantes de Fusión/efectos adversos , Atrofias Musculares Espinales de la Infancia/terapia , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Autopsia , Productos Biológicos/administración & dosificación , ADN/genética , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Humanos , Lactante , Recién Nacido , Masculino , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Atrofias Musculares Espinales de la Infancia/genética , Atrofias Musculares Espinales de la Infancia/mortalidad , Atrofias Musculares Espinales de la Infancia/patología , Distribución Tisular/efectos de los fármacos
12.
Blood ; 138(22): 2256-2268, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34587242

RESUMEN

SARS-CoV-2 vaccine ChAdOx1 nCoV-19 (AstraZeneca) causes a thromboembolic complication termed vaccine-induced immune thrombotic thrombocytopenia (VITT). Using biophysical techniques, mouse models, and analysis of VITT patient samples, we identified determinants of this vaccine-induced adverse reaction. Super-resolution microscopy visualized vaccine components forming antigenic complexes with platelet factor 4 (PF4) on platelet surfaces to which anti-PF4 antibodies obtained from VITT patients bound. PF4/vaccine complex formation was charge-driven and increased by addition of DNA. Proteomics identified substantial amounts of virus production-derived T-REx HEK293 proteins in the ethylenediaminetetraacetic acid (EDTA)-containing vaccine. Injected vaccine increased vascular leakage in mice, leading to systemic dissemination of vaccine components known to stimulate immune responses. Together, PF4/vaccine complex formation and the vaccine-stimulated proinflammatory milieu trigger a pronounced B-cell response that results in the formation of high-avidity anti-PF4 antibodies in VITT patients. The resulting high-titer anti-PF4 antibodies potently activated platelets in the presence of PF4 or DNA and polyphosphate polyanions. Anti-PF4 VITT patient antibodies also stimulated neutrophils to release neutrophil extracellular traps (NETs) in a platelet PF4-dependent manner. Biomarkers of procoagulant NETs were elevated in VITT patient serum, and NETs were visualized in abundance by immunohistochemistry in cerebral vein thrombi obtained from VITT patients. Together, vaccine-induced PF4/adenovirus aggregates and proinflammatory reactions stimulate pathologic anti-PF4 antibody production that drives thrombosis in VITT. The data support a 2-step mechanism underlying VITT that resembles the pathogenesis of (autoimmune) heparin-induced thrombocytopenia.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Autoanticuerpos/inmunología , COVID-19/prevención & control , Proteínas de la Cápside/efectos adversos , ChAdOx1 nCoV-19/efectos adversos , Contaminación de Medicamentos , Vectores Genéticos/efectos adversos , Células HEK293/inmunología , Inmunoglobulina G/inmunología , Factor Plaquetario 4/inmunología , Púrpura Trombocitopénica Idiopática/etiología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/efectos adversos , Adenoviridae/inmunología , Animales , Complejo Antígeno-Anticuerpo/ultraestructura , Autoanticuerpos/biosíntesis , Síndrome de Fuga Capilar/etiología , Proteínas de la Cápside/inmunología , Línea Celular Transformada , ChAdOx1 nCoV-19/química , ChAdOx1 nCoV-19/inmunología , ChAdOx1 nCoV-19/toxicidad , Dispersión Dinámica de Luz , Epítopos/química , Epítopos/inmunología , Trampas Extracelulares/inmunología , Extravasación de Materiales Terapéuticos y Diagnósticos/etiología , Vectores Genéticos/inmunología , Células HEK293/química , Humanos , Imagenología Tridimensional , Inmunoglobulina G/biosíntesis , Inflamación , Ratones , Microscopía/métodos , Activación Plaquetaria , Proteómica , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/inmunología , Trombosis de los Senos Intracraneales/diagnóstico por imagen , Trombosis de los Senos Intracraneales/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Cultivo de Virus
13.
Int J Lab Hematol ; 43 Suppl 1: 117-123, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34288447

RESUMEN

Since the cloning and characterization of the factor VIII (FVIII) and factor IX genes in the mid-1980s, gene therapy has been perceived as having significant potential for the treatment of severe hemophilia. Now, some 35 years later, these proposals are close to being realized through the licensing of the first clinical gene therapy product. Adeno-associated viral vector-mediated gene therapy for hemophilia A and B has been extensively investigated in preclinical models over the past 20 years, and since 2011, there has been increasing evidence in early phase clinical trials that this therapeutic strategy can provide safe and effective rescue of the hemostatic phenotype in severe hemophilia. As the uptake of hemophilia gene therapy progresses, it is clear that many aspects of the gene therapy process require crucial laboratory support to ensure safe and effective outcomes from his new therapeutic paradigm. These laboratory contributions extend from evaluations of the gene therapy vehicle, assessments of the patient immune status for the vector, and ultimately the performance of assays to determine the hemostatic benefit of the gene therapy and potentially of its long-term safety on the host genome. As with many aspects of past hemophilia care, the safe and effective delivery of gene therapy will require an informed and coordinated contribution from laboratory science.


Asunto(s)
Terapia Genética , Hemofilia A/terapia , Hemofilia B/terapia , Animales , Coagulación Sanguínea , Pruebas de Coagulación Sanguínea , Ensayos Clínicos como Asunto , Dependovirus/genética , Factor IX/genética , Factor VIII/genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Hemofilia A/sangre , Hemofilia A/diagnóstico , Hemofilia A/genética , Hemofilia B/sangre , Hemofilia B/diagnóstico , Hemofilia B/genética , Humanos , Mutación , Fenotipo , Resultado del Tratamiento
14.
Mol Ther ; 29(12): 3383-3397, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34174440

RESUMEN

Hematopoietic stem cell gene therapy is emerging as a promising therapeutic strategy for many diseases of the blood and immune system. However, several individuals who underwent gene therapy in different trials developed hematological malignancies caused by insertional mutagenesis. Preclinical assessment of vector safety remains challenging because there are few reliable assays to screen for potential insertional mutagenesis effects in vitro. Here we demonstrate that genotoxic vectors induce a unique gene expression signature linked to stemness and oncogenesis in transduced murine hematopoietic stem and progenitor cells. Based on this finding, we developed the surrogate assay for genotoxicity assessment (SAGA). SAGA classifies integrating retroviral vectors using machine learning to detect this gene expression signature during the course of in vitro immortalization. On a set of benchmark vectors with known genotoxic potential, SAGA achieved an accuracy of 90.9%. SAGA is more robust and sensitive and faster than previous assays and reliably predicts a mutagenic risk for vectors that led to leukemic severe adverse events in clinical trials. Our work provides a fast and robust tool for preclinical risk assessment of gene therapy vectors, potentially paving the way for safer gene therapy trials.


Asunto(s)
Terapia Genética , Vectores Genéticos , Animales , Daño del ADN , Expresión Génica , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Células Madre Hematopoyéticas , Humanos , Aprendizaje Automático , Ratones , Mutagénesis Insercional
15.
Int J Mol Sci ; 22(11)2021 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-34070997

RESUMEN

Mucopolysaccharidosis type II (MPS II) is a lysosomal storage disorder based on a mutation in the IDS gene that encodes iduronate 2-sulphatase. As a result, there is an accumulation of glycosaminoglycans-heparan sulphate and dermatan sulphate-in almost all body tissues, which leads to their dysfunction. Currently, the primary treatment is enzyme replacement therapy, which improves the course of the disease by reducing somatic symptoms, including hepatomegaly and splenomegaly. The enzyme, however, does not cross the blood-brain barrier, and no improvement in the function of the central nervous system has been observed in patients with the severe form of the disease. An alternative method of treatment that solves typical problems of enzyme replacement therapy is gene therapy, i.e., delivery of the correct gene to target cells through an appropriate vector. Much progress has been made in applying gene therapy for MPS II, from cellular models to human clinical trials. In this article, we briefly present the history and basics of gene therapy and discuss the current state of knowledge about the methods of this therapy in mucopolysaccharidosis type II.


Asunto(s)
Glicoproteínas/genética , Mucopolisacaridosis II/terapia , Adolescente , Animales , Barrera Hematoencefálica , Sistemas CRISPR-Cas , Niño , Preescolar , Ensayos Clínicos como Asunto , Dependovirus/genética , Modelos Animales de Enfermedad , Portadores de Fármacos , Electroporación , Terapia de Reemplazo Enzimático/métodos , Edición Génica , Terapia Genética , Vectores Genéticos/efectos adversos , Vectores Genéticos/uso terapéutico , Glicoproteínas/farmacocinética , Glicoproteínas/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/uso terapéutico , Lactante , Inyecciones Intraventriculares , Inyecciones Espinales , Lentivirus/genética , Ratones , Mucopolisacaridosis II/genética , Estudios Multicéntricos como Asunto , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Retroviridae/genética , Nucleasas de los Efectores Tipo Activadores de la Transcripción
16.
Front Immunol ; 12: 655478, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34040605

RESUMEN

Mucopolysaccharidosis type IIIB syndrome (Sanfilippo disease) is a rare autosomic recessif disorder caused by mutations in the α-N-acetylglucosaminidase (NAGLU) gene coding for a lysosomal enzyme, leading to neurodegeneration and progressive deterioration of cognitive abilities in affected children. To supply the missing enzyme, several recent human gene therapy trials relied on the deposit of adeno-associated virus (AAV) vectors directly into the brain. We reported safety and efficacy of an intracerebral therapy in a phase 1/2 clinical trial (https://clinicaltrials.gov/ct2/show/NCT03300453), with a recombinant AAV serotype 2/5 (rAAV2/5) coding human NAGLU in four children with MPS IIIB syndrome receiving immunosuppression. It was reported that AAV-mediated gene therapies might elicit a strong host immune response resulting in decreased transgene expression. To address this issue, we performed a comprehensive analysis of cellular immunity and cytokine patterns generated against the therapeutic enzyme in the four treated children over 5.5 years of follow-up. We report the emergence of memory and polyfunctional CD4+ and CD8+ T lymphocytes sensitized to the transgene soon after the start of therapy, and appearing in peripheral blood in waves throughout the follow-up. However, this response had no apparent impact on CNS transgene expression, which remained stable 66 months after surgery, possibly a consequence of the long-term immunosuppressive treatment. We also report that gene therapy did not trigger neuroinflammation, evaluated through the expression of cytokines and chemokines in patients' CSF. Milder disease progression in the youngest patient was found associated with low level and less differentiated circulating NAGLU-specific T cells, together with the lack of proinflammatory cytokines in the CSF. Findings in this study support a systematic and comprehensive immunomonitoring approach for understanding the impact immune reactions might have on treatment safety and efficacy of gene therapies.


Asunto(s)
Acetilglucosaminidasa/inmunología , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Inmunidad Celular , Mucopolisacaridosis III/complicaciones , Transgenes/inmunología , Acetilglucosaminidasa/genética , Niño , Citocinas/metabolismo , Vías de Administración de Medicamentos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Memoria Inmunológica , Activación de Linfocitos , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/terapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transgenes/genética
17.
J Mol Med (Berl) ; 99(8): 1057-1071, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34021360

RESUMEN

Gene therapy of genetically determined diseases, including some pathologies of the respiratory system, requires an efficient method for transgene delivery. Recombinant adeno-associated viral (rAAV) vectors are well studied and employed in gene therapy, as they are relatively simple and low immunogenic and able to efficiently transduce eukaryotic cells. To date, many natural and artificial (with modified capsids) AAV serotypes have been isolated, demonstrating preferential tropism toward different tissues and cells in accordance with the prevalent receptors on the cell surface. However, rAAV-mediated delivery is not strictly specific due to wide tropism of some viral serotypes. Thus, the development of the methods allowing modulating specificity of these vectors could be beneficial in some cases. This review describes various approaches for retargeting rAAV to respiratory cells, for example, using different types of capsid modifications and regulation of a transgene expression by tissue-specific promoters. Part of the review is devoted to the issues of transduction of stem and progenitor lung cells using AAV, which is a complicated task today.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/terapia , Transducción Genética , Animales , Dependovirus/clasificación , Dependovirus/ultraestructura , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Regulación de la Expresión Génica , Ingeniería Genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Especificidad de Órganos/genética , Regiones Promotoras Genéticas , Células Madre/metabolismo , Transgenes
18.
Mol Pharm ; 18(6): 2142-2160, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34010004

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease, for which no effective treatment is yet available to either slow or terminate it. Recent advances in gene therapy renew hope for developing an effective approach to control this disease. Non-viral vectors, such as lipid- and polymer-based nanoparticles, cationic polymers, and exosomes, can effectively transfer genes into primary neurons. The resulting gene expression can be long-term, stable, and without immunological complications, which is essential for the effective management of neurological disorders. This Review will first describe the current research and clinical stage of novel therapies for ALS. It will then touch on the journey of non-viral vector use in ALS, subsequently highlighting the application of non-viral vector-mediated gene therapy. The bottlenecks in the translation of non-viral vectors for ALS treatment are also discussed, including the biological barriers of systemic administration and the issues of "when, where, and how much?" for effective gene delivery. The prospect of employing emerging techniques, such as CRISPR-Cas9 gene editing, stem cell methodology, and low-intensity focused ultrasound for fueling the transport of non-viral vectors to the central nervous system for personalized gene therapy, is briefly discussed in the context of ALS. Despite the challenging road that lies ahead, with the current expansion in interest and technological advancement in non-viral vector-delivered gene therapy for ALS, we hold hope that the field is headed toward a positive future.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Técnicas de Transferencia de Gen/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Esclerosis Amiotrófica Lateral/genética , Animales , Sistemas CRISPR-Cas/genética , Células Cultivadas , Ensayos Clínicos Fase III como Asunto , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen/tendencias , Terapia Genética/efectos adversos , Terapia Genética/tendencias , Vectores Genéticos/efectos adversos , Humanos , Nanopartículas/administración & dosificación , Cultivo Primario de Células
19.
Front Immunol ; 12: 658399, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815421

RESUMEN

Adeno-associated virus (AAV) vector-based gene therapy is currently the only in vivo gene therapy approved in the US and Europe. The recent tragic death of three children in a clinical trial to treat X-Linked Myotubular Myopathy by delivering myotubularin with an AAV8 vector notwithstanding, AAV remains a highly promising therapeutic gene delivery platform. But the successful use of AAV vectors to treat an increasing number of diseases also makes establishing protocols to determine therapeutically relevant titers of pre-existing anti-AAV antibodies and approaches to deplete those antibodies more urgent than ever. In this mini review, I will briefly discuss (i) our knowledge regarding the prevalence of anti-AAV antibodies, (ii) the challenges to measure those antibodies by methods that are most predictive of their influence on therapeutic efficacy of AAV gene transfer, and (iii) approaches to overcome the formidable hurdle that anti-AAV antibodies pose to the successful clinical use of AAV gene therapy.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Dependovirus/inmunología , Vectores Genéticos/efectos adversos , Animales , Bioensayo , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Humanos
20.
Front Immunol ; 12: 610042, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679747

RESUMEN

Adoptive cell therapy (ACT) using autologous tumor infiltrating lymphocytes (TIL) achieves durable clinical benefit for patients from whom these cells can be derived in advanced metastatic melanoma but is limited in most solid tumors as a result of immune escape and exclusion. A tumor microenvironment (TME) priming strategy to improve the quantity and quality of TIL represents an important tactic to explore. Oncolytic viruses expressing immune stimulatory cytokines induce a potent inflammatory response that may enhance infiltration and activation of T cells. In this study, we examined the ability of an attenuated oncolytic vaccinia virus expressing IL15/IL15Rα (vvDD-IL15/Rα) to enhance recovery of lavage T cells in peritoneal carcinomatosis (PC). We found that intraperitoneal (IP) vvDD-IL15/Rα treatment of animals bearing PC resulted in a significant increase in cytotoxic function and memory formation in CD8+ T cells in peritoneal fluid. Using tetramers for vaccinia virus B8R antigen and tumor rejection antigen p15E, we found that the expanded population of peritoneal CD8+ T cells are specific for vaccinia or tumor with increased tumor-specificity over time, reinforced with viral clearance. Application of these vvDD-IL15/Rα induced CD8+ T cells in ACT of a lethal model of PC significantly increased survival. In addition, we found in patients with peritoneal metastases from various primary solid tumors that peritoneal T cells could be recovered but were exhausted with infrequent tumor-reactivity. If clinically translatable, vvDD-IL15/Rα in vivo priming would greatly expand the number of patients with advanced metastatic cancers responsive to T cell therapy.


Asunto(s)
Vectores Genéticos/efectos adversos , Inmunoterapia Adoptiva , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Virus Oncolíticos/inmunología , Neoplasias Peritoneales/terapia , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Interleucina-15/metabolismo , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Ratones , Viroterapia Oncolítica/efectos adversos , Virus Oncolíticos/genética , Neoplasias Peritoneales/secundario , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...