Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.217
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 185(8): 1261-1265, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35385685

RESUMEN

Through studies in mice and in humans, Stuart Orkin showed that GATA-1 is a master transcriptional regulator of hematopoiesis. He has highlighted the role of BCL11A in the fetal-adult hemoglobin switch. The Gairdner Foundation Award recognizes Orkin's contribution to the development of gene therapy of sickle cell disease.


Asunto(s)
Anemia de Células Falciformes , Distinciones y Premios , Terapia Genética , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Animales , Modelos Animales de Enfermedad , Hemoglobina Fetal/genética , Hematopoyesis/genética , Humanos , Ratones , Proteínas Represoras/genética
2.
N Engl J Med ; 390(18): 1649-1662, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38661449

RESUMEN

BACKGROUND: Exagamglogene autotemcel (exa-cel) is a nonviral cell therapy designed to reactivate fetal hemoglobin synthesis by means of ex vivo clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing of autologous CD34+ hematopoietic stem and progenitor cells (HSPCs) at the erythroid-specific enhancer region of BCL11A. METHODS: We conducted a phase 3, single-group, open-label study of exa-cel in patients 12 to 35 years of age with sickle cell disease who had had at least two severe vaso-occlusive crises in each of the 2 years before screening. CD34+ HSPCs were edited with the use of CRISPR-Cas9. Before the exa-cel infusion, patients underwent myeloablative conditioning with pharmacokinetically dose-adjusted busulfan. The primary end point was freedom from severe vaso-occlusive crises for at least 12 consecutive months. A key secondary end point was freedom from inpatient hospitalization for severe vaso-occlusive crises for at least 12 consecutive months. The safety of exa-cel was also assessed. RESULTS: A total of 44 patients received exa-cel, and the median follow-up was 19.3 months (range, 0.8 to 48.1). Neutrophils and platelets engrafted in each patient. Of the 30 patients who had sufficient follow-up to be evaluated, 29 (97%; 95% confidence interval [CI], 83 to 100) were free from vaso-occlusive crises for at least 12 consecutive months, and all 30 (100%; 95% CI, 88 to 100) were free from hospitalizations for vaso-occlusive crises for at least 12 consecutive months (P<0.001 for both comparisons against the null hypothesis of a 50% response). The safety profile of exa-cel was generally consistent with that of myeloablative busulfan conditioning and autologous HSPC transplantation. No cancers occurred. CONCLUSIONS: Treatment with exa-cel eliminated vaso-occlusive crises in 97% of patients with sickle cell disease for a period of 12 months or more. (CLIMB SCD-121; ClinicalTrials.gov number, NCT03745287.).


Asunto(s)
Anemia de Células Falciformes , Hemoglobina Fetal , Trasplante de Células Madre Hematopoyéticas , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Adulto Joven , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Antígenos CD34 , Busulfano/uso terapéutico , Sistemas CRISPR-Cas , Hemoglobina Fetal/biosíntesis , Hemoglobina Fetal/genética , Edición Génica , Células Madre Hematopoyéticas , Proteínas Represoras , Acondicionamiento Pretrasplante , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Agonistas Mieloablativos/uso terapéutico , Europa (Continente) , América del Norte
3.
Nature ; 595(7866): 295-302, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34079130

RESUMEN

Sickle cell disease (SCD) is caused by a mutation in the ß-globin gene HBB1. We used a custom adenine base editor (ABE8e-NRCH)2,3 to convert the SCD allele (HBBS) into Makassar ß-globin (HBBG), a non-pathogenic variant4,5. Ex vivo delivery of mRNA encoding the base editor with a targeting guide RNA into haematopoietic stem and progenitor cells (HSPCs) from patients with SCD resulted in 80% conversion of HBBS to HBBG. Sixteen weeks after transplantation of edited human HSPCs into immunodeficient mice, the frequency of HBBG was 68% and hypoxia-induced sickling of bone marrow reticulocytes had decreased fivefold, indicating durable gene editing. To assess the physiological effects of HBBS base editing, we delivered ABE8e-NRCH and guide RNA into HSPCs from a humanized SCD mouse6 and then transplanted these cells into irradiated mice. After sixteen weeks, Makassar ß-globin represented 79% of ß-globin protein in blood, and hypoxia-induced sickling was reduced threefold. Mice that received base-edited HSPCs showed near-normal haematological parameters and reduced splenic pathology compared to mice that received unedited cells. Secondary transplantation of edited bone marrow confirmed that the gene editing was durable in long-term haematopoietic stem cells and showed that HBBS-to-HBBG editing of 20% or more is sufficient for phenotypic rescue. Base editing of human HSPCs avoided the p53 activation and larger deletions that have been observed following Cas9 nuclease treatment. These findings point towards a one-time autologous treatment for SCD that eliminates pathogenic HBBS, generates benign HBBG, and minimizes the undesired consequences of double-strand DNA breaks.


Asunto(s)
Adenina/metabolismo , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Edición Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Globinas beta/genética , Animales , Antígenos CD34/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Genoma Humano/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/patología , Humanos , Masculino , Ratones
4.
Annu Rev Genomics Hum Genet ; 24: 255-275, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37624668

RESUMEN

Sickle cell disease (SCD) is a monogenic blood disease caused by a point mutation in the gene coding for ß-globin. The abnormal hemoglobin [sickle hemoglobin (HbS)] polymerizes under low-oxygen conditions and causes red blood cells to sickle. The clinical presentation varies from very severe (with acute pain, chronic pain, and early mortality) to normal (few complications and a normal life span). The variability of SCD might be due (in part) to various genetic modulators. First, we review the main genetic factors, polymorphisms, and modifier genes that influence the expression of globin or otherwise modulate the severity of SCD. Considering SCD as a complex, multifactorial disorder is important for the development of appropriate pharmacological and genetic treatments. Second, we review the characteristics, advantages, and disadvantages of the latest advances in gene therapy for SCD, from lentiviral-vector-based approaches to gene-editing strategies.


Asunto(s)
Dolor Agudo , Anemia de Células Falciformes , Dolor Crónico , Hemoglobinas Anormales , Humanos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Eritrocitos
5.
N Engl J Med ; 389(9): 820-832, 2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37646679

RESUMEN

BACKGROUND: Sickle cell disease is caused by a defect in the ß-globin subunit of adult hemoglobin. Sickle hemoglobin polymerizes under hypoxic conditions, producing deformed red cells that hemolyze and cause vaso-occlusion that results in progressive organ damage and early death. Elevated fetal hemoglobin levels in red cells protect against complications of sickle cell disease. OTQ923, a clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-edited CD34+ hematopoietic stem- and progenitor-cell (HSPC) product, has a targeted disruption of the HBG1 and HBG2 (γ-globin) gene promoters that increases fetal hemoglobin expression in red-cell progeny. METHODS: We performed a tiling CRISPR-Cas9 screen of the HBG1 and HBG2 promoters by electroporating CD34+ cells obtained from healthy donors with Cas9 complexed with one of 72 guide RNAs, and we assessed the fraction of fetal hemoglobin-immunostaining erythroblasts (F cells) in erythroid-differentiated progeny. The gRNA resulting in the highest level of F cells (gRNA-68) was selected for clinical development. We enrolled participants with severe sickle cell disease in a multicenter, phase 1-2 clinical study to assess the safety and adverse-effect profile of OTQ923. RESULTS: In preclinical experiments, CD34+ HSPCs (obtained from healthy donors and persons with sickle cell disease) edited with CRISPR-Cas9 and gRNA-68 had sustained on-target editing with no off-target mutations and produced high levels of fetal hemoglobin after in vitro differentiation or xenotransplantation into immunodeficient mice. In the study, three participants received autologous OTQ923 after myeloablative conditioning and were followed for 6 to 18 months. At the end of the follow-up period, all the participants had engraftment and stable induction of fetal hemoglobin (fetal hemoglobin as a percentage of total hemoglobin, 19.0 to 26.8%), with fetal hemoglobin broadly distributed in red cells (F cells as a percentage of red cells, 69.7 to 87.8%). Manifestations of sickle cell disease decreased during the follow-up period. CONCLUSIONS: CRISPR-Cas9 disruption of the HBG1 and HBG2 gene promoters was an effective strategy for induction of fetal hemoglobin. Infusion of autologous OTQ923 into three participants with severe sickle cell disease resulted in sustained induction of red-cell fetal hemoglobin and clinical improvement in disease severity. (Funded by Novartis Pharmaceuticals; ClinicalTrials.gov number, NCT04443907.).


Asunto(s)
Anemia de Células Falciformes , Sistemas CRISPR-Cas , Eritrocitos , Hemoglobina Fetal , Trasplante de Células Madre Hematopoyéticas , Animales , Ratones , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Antígenos CD34 , Hemoglobina Fetal/biosíntesis , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Hemoglobina Falciforme , Regiones Promotoras Genéticas
6.
Blood ; 143(11): 967-970, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38289232

RESUMEN

ABSTRACT: The root cause of sickle cell anemia has been known for 7 decades, yet no curative therapies have been available other than allogeneic bone marrow transplantation, for which applicability is limited. Two potentially curative therapies based on gene therapy and gene editing strategies have recently received US Food and Drug Administration approval. This review surveys the nature of these therapies and the opportunities and issues raised by the prospect of definitive genetically based therapies being available in clinical practice.


Asunto(s)
Anemia de Células Falciformes , Trasplante de Células Madre Hematopoyéticas , Humanos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Trasplante de Médula Ósea , Edición Génica , Terapia Genética/métodos
7.
Blood ; 143(9): 769-776, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-37979134

RESUMEN

ABSTRACT: Fifty years ago, people with sickle cell disease (SCD) were discouraged from becoming pregnant, but now, most should be supported if they choose to pursue a pregnancy. They and their providers, however, should be aware of the physiological changes of pregnancy that aggravate SCD and pregnancy's unique maternal and fetal challenges. Maternal problems can arise from chronic underlying organ dysfunction such as renal disease or pulmonary hypertension; from acute complications of SCD such as acute anemia, vaso-occlusive crises, and acute chest syndrome; and/or from pregnancy-related complications such as preeclampsia, sepsis, severe anemia, thromboembolism, and the need for cesarean delivery. Fetal problems include alloimmunization, opioid exposure, fetal growth restriction, preterm delivery, and stillbirth. Before and during pregnancy, in addition to the assessment and care that every pregnant patient should receive, patients with SCD should be evaluated and treated by a multidisciplinary team with respect to their unique maternal and fetal issues.


Asunto(s)
Síndrome Torácico Agudo , Anemia de Células Falciformes , Preeclampsia , Complicaciones del Embarazo , Embarazo , Recién Nacido , Femenino , Humanos , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/terapia , Síndrome Torácico Agudo/etiología , Complicaciones del Embarazo/terapia , Atención Prenatal
8.
Blood ; 143(25): 2654-2665, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38493482

RESUMEN

ABSTRACT: In the setting of a learning collaborative, we conducted an international multicenter phase 2 clinical trial testing the hypothesis that nonmyeloablative-related haploidentical bone marrow transplant (BMT) with thiotepa and posttransplant cyclophosphamide (PTCy) will result in 2-year event-free survival (no graft failure or death) of at least 80%. A total of 70 participants were evaluable based on the conditioning protocol. Graft failure occurred in 8 of 70 (11.4%) and only in participants aged <18 years; all had autologous reconstitution. After a median follow-up of 2.4 years, the 2-year Kaplan-Meier-based probability of event-free survival was 82.6%. The 2-year overall survival was 94.1%, with no difference between children and adult participants. After excluding participants with graft failure (n = 8), participants with engraftment had median whole blood donor chimerism values at days +180 and +365 after transplant of 100% (n = 58), respectively, and 96.6% (57/59) were off immunosuppression 1 year after transplant. The 1-year grade 3 to 4 acute graft-versus-host disease (GVHD) rate was 10%, and the 2-year moderate-severe chronic GVHD rate was 10%. Five participants (7.1%) died from infectious complications. We demonstrate that nonmyeloablative haploidentical BMT with thiotepa and PTCy is a readily available curative therapy for most adults, even those with organ damage, compared to the more expensive myeloablative gene therapy and gene editing. Additional strategies are required for children to decrease graft failure rates. The trial was registered at www.clinicaltrials.gov as #NCT01850108.


Asunto(s)
Anemia de Células Falciformes , Trasplante de Médula Ósea , Enfermedad Injerto contra Huésped , Trasplante Haploidéntico , Humanos , Trasplante de Médula Ósea/métodos , Trasplante de Médula Ósea/efectos adversos , Masculino , Femenino , Niño , Adolescente , Adulto , Anemia de Células Falciformes/terapia , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante Haploidéntico/métodos , Preescolar , Adulto Joven , Ciclofosfamida/uso terapéutico , Ciclofosfamida/administración & dosificación , Acondicionamiento Pretrasplante/métodos , Persona de Mediana Edad , Tiotepa/administración & dosificación , Tiotepa/uso terapéutico
9.
Nature ; 578(7794): 229-236, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32051598

RESUMEN

Genome editing, which involves the precise manipulation of cellular DNA sequences to alter cell fates and organism traits, has the potential to both improve our understanding of human genetics and cure genetic disease. Here I discuss the scientific, technical and ethical aspects of using CRISPR (clustered regularly interspaced short palindromic repeats) technology for therapeutic applications in humans, focusing on specific examples that highlight both opportunities and challenges. Genome editing is-or will soon be-in the clinic for several diseases, with more applications under development. The rapid pace of the field demands active efforts to ensure that this breakthrough technology is used responsibly to treat, cure and prevent genetic disease.


Asunto(s)
Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Edición Génica/métodos , Edición Génica/tendencias , Genoma Humano/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Sistemas CRISPR-Cas/genética , Edición Génica/ética , Edición Génica/normas , Mutación de Línea Germinal/genética , Humanos , Especificidad de Órganos/genética , Seguridad del Paciente , Globinas beta/genética
10.
Trends Genet ; 38(12): 1284-1298, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35934593

RESUMEN

Sickle cell disease (SCD) is a common genetic blood disorder associated with acute and chronic pain, progressive multiorgan damage, and early mortality. Recent advances in technologies to manipulate the human genome, a century of research and the development of techniques enabling the isolation, efficient genetic modification, and reimplantation of autologous patient hematopoietic stem cells (HSCs), mean that curing most patients with SCD could soon be a reality in wealthy countries. In parallel, ongoing research is pursuing more facile treatments, such as in-vivo-delivered genetic therapies and new drugs that can eventually be administered in low- and middle-income countries where most SCD patients reside.


Asunto(s)
Anemia de Células Falciformes , Trasplante de Células Madre Hematopoyéticas , Humanos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Edición Génica/métodos , Células Madre Hematopoyéticas , Terapia Genética
11.
Annu Rev Med ; 74: 473-487, 2023 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-36067800

RESUMEN

Sickle cell disease (SCD) results from a single base pair change in the sixth codon of the ß-globin chain of hemoglobin, which promotes aggregation of deoxyhemoglobin, increasing rigidity of red blood cells and causing vaso-occlusive and hemolytic complications. Allogeneic transplant of hematopoietic stem cells (HSCs) can eliminate SCD manifestations but is limited by absence of well-matched donors and immune complications. Gene therapy with transplantation of autologous HSCs that are gene-modified may provide similar benefits without the immune complications. Much progress has been made, and patients are realizing significant clinical improvements in multiple trials using different approaches with lentiviral vector-mediated gene addition to inhibit hemoglobin aggregation. Gene editing approaches are under development to provide additional therapeutic opportunities. Gene therapy for SCD has advanced from an attractive concept to clinical reality.


Asunto(s)
Anemia de Células Falciformes , Trasplante de Células Madre Hematopoyéticas , Humanos , Trasplante de Células Madre Hematopoyéticas/métodos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Terapia Genética/métodos , Células Madre Hematopoyéticas , Hemoglobinas/genética
12.
Am J Hum Genet ; 109(1): 180-191, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34968422

RESUMEN

Next-generation sequencing (NGS) technologies have transformed medical genetics. However, short-read lengths pose a limitation on identification of structural variants, sequencing repetitive regions, phasing of distant nucleotide changes, and distinguishing highly homologous genomic regions. Long-read sequencing technologies may offer improvements in the characterization of genes that are currently difficult to assess. We used a combination of targeted DNA capture, long-read sequencing, and a customized bioinformatics pipeline to fully assemble the RH region, which harbors variation relevant to red cell donor-recipient mismatch, particularly among patients with sickle cell disease. RHD and RHCE are a pair of duplicated genes located within an ∼175 kb region on human chromosome 1 that have high sequence similarity and frequent structural variations. To achieve the assembly, we utilized palindrome repeats in PacBio SMRT reads to obtain consensus sequences of 2.1 to 2.9 kb average length with over 99% accuracy. We used these long consensus sequences to identify 771 assembly markers and to phase the RHD-RHCE region with high confidence. The dataset enabled direct linkage between coding and intronic variants, phasing of distant SNPs to determine RHD-RHCE haplotypes, and identification of known and novel structural variations along with the breakpoints. A limiting factor in phasing is the frequency of heterozygous assembly markers and therefore was most successful in samples from African Black individuals with increased heterogeneity at the RH locus. Overall, this approach allows RH genotyping and de novo assembly in an unbiased and comprehensive manner that is necessary to expand application of NGS technology to high-resolution RH typing.


Asunto(s)
Transfusión Sanguínea , Duplicación de Gen , Variación Genética , Sistema del Grupo Sanguíneo Rh-Hr/genética , Alelos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Rotura Cromosómica , Biología Computacional/métodos , Frecuencia de los Genes , Heterogeneidad Genética , Ligamiento Genético , Genómica/métodos , Haplotipos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Polimorfismo Genético , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN/métodos
13.
N Engl J Med ; 386(7): 617-628, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34898139

RESUMEN

BACKGROUND: Sickle cell disease is characterized by the painful recurrence of vaso-occlusive events. Gene therapy with the use of LentiGlobin for sickle cell disease (bb1111; lovotibeglogene autotemcel) consists of autologous transplantation of hematopoietic stem and progenitor cells transduced with the BB305 lentiviral vector encoding a modified ß-globin gene, which produces an antisickling hemoglobin, HbAT87Q. METHODS: In this ongoing phase 1-2 study, we optimized the treatment process in the initial 7 patients in Group A and 2 patients in Group B with sickle cell disease. Group C was established for the pivotal evaluation of LentiGlobin for sickle cell disease, and we adopted a more stringent inclusion criterion that required a minimum of four severe vaso-occlusive events in the 24 months before enrollment. In this unprespecified interim analysis, we evaluated the safety and efficacy of LentiGlobin in 35 patients enrolled in Group C. Included in this analysis was the number of severe vaso-occlusive events after LentiGlobin infusion among patients with at least four vaso-occlusive events in the 24 months before enrollment and with at least 6 months of follow-up. RESULTS: As of February 2021, cell collection had been initiated in 43 patients in Group C; 35 received a LentiGlobin infusion, with a median follow-up of 17.3 months (range, 3.7 to 37.6). Engraftment occurred in all 35 patients. The median total hemoglobin level increased from 8.5 g per deciliter at baseline to 11 g or more per deciliter from 6 months through 36 months after infusion. HbAT87Q contributed at least 40% of total hemoglobin and was distributed across a mean (±SD) of 85±8% of red cells. Hemolysis markers were reduced. Among the 25 patients who could be evaluated, all had resolution of severe vaso-occlusive events, as compared with a median of 3.5 events per year (range, 2.0 to 13.5) in the 24 months before enrollment. Three patients had a nonserious adverse event related or possibly related to LentiGlobin that resolved within 1 week after onset. No cases of hematologic cancer were observed during up to 37.6 months of follow-up. CONCLUSIONS: One-time treatment with LentiGlobin resulted in sustained production of HbAT87Q in most red cells, leading to reduced hemolysis and complete resolution of severe vaso-occlusive events. (Funded by Bluebird Bio; HGB-206 ClinicalTrials.gov number, NCT02140554.).


Asunto(s)
Anemia de Células Falciformes/terapia , Terapia Genética , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Hemoglobinas/genética , Lentivirus , Trasplante de Células Madre , Globinas beta/genética , Adolescente , Adulto , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/complicaciones , Niño , Femenino , Hemoglobina Fetal , Hemoglobinas/análisis , Hemoglobinas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Grado de Desobstrucción Vascular , Adulto Joven
14.
N Engl J Med ; 386(2): 138-147, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34898140

RESUMEN

Gene therapy with LentiGlobin for sickle cell disease (bb1111, lovotibeglogene autotemcel) consists of autologous transplantation of a patient's hematopoietic stem cells transduced with the BB305 lentiviral vector that encodes the ßA-T87Q-globin gene. Acute myeloid leukemia developed in a woman approximately 5.5 years after she had received LentiGlobin for sickle cell disease as part of the initial cohort (Group A) of the HGB-206 study. An analysis of peripheral-blood samples revealed that blast cells contained a BB305 lentiviral vector insertion site. The results of an investigation of causality indicated that the leukemia was unlikely to be related to vector insertion, given the location of the insertion site, the very low transgene expression in blast cells, and the lack of an effect on expression of surrounding genes. Several somatic mutations predisposing to acute myeloid leukemia were present after diagnosis, which suggests that patients with sickle cell disease are at increased risk for hematologic malignant conditions after transplantation, most likely because of a combination of risks associated with underlying sickle cell disease, transplantation procedure, and inadequate disease control after treatment. (Funded by Bluebird Bio.).


Asunto(s)
Anemia de Células Falciformes/terapia , Expresión Génica , Terapia Genética/efectos adversos , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/etiología , Globinas beta/genética , Adulto , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Carcinogénesis , Femenino , Vectores Genéticos , Humanos , Lentivirus , Factores de Riesgo , Análisis de Secuencia de ARN , Transgenes , Trasplante Autólogo
15.
Blood ; 141(17): 2085-2099, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-36800642

RESUMEN

Sickle cell disease (SCD) is a monogenic disease caused by a nucleotide mutation in the ß-globin gene. Current gene therapy studies are mainly focused on lentiviral vector-mediated gene addition or CRISPR/Cas9-mediated fetal globin reactivation, leaving the root cause unfixed. We developed a vectorized prime editing system that can directly repair the SCD mutation in hematopoietic stem cells (HSCs) in vivo in a SCD mouse model (CD46/Townes mice). Our approach involved a single intravenous injection of a nonintegrating, prime editor-expressing viral vector into mobilized CD46/Townes mice and low-dose drug selection in vivo. This procedure resulted in the correction of ∼40% of ßS alleles in HSCs. On average, 43% of sickle hemoglobin was replaced by adult hemoglobin, thereby greatly mitigating the SCD phenotypes. Transplantation in secondary recipients demonstrated that long-term repopulating HSCs were edited. Highly efficient target site editing was achieved with minimal generation of insertions and deletions and no detectable off-target editing. Because of its simplicity and portability, our in vivo prime editing approach has the potential for application in resource-poor countries where SCD is prevalent.


Asunto(s)
Anemia de Células Falciformes , Edición Génica , Ratones , Animales , Edición Génica/métodos , Sistemas CRISPR-Cas , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Células Madre Hematopoyéticas , Hemoglobina Falciforme/genética
16.
Blood ; 141(4): 335-344, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36040484

RESUMEN

Children with sickle cell disease (SCD) demonstrate cerebral hemodynamic stress and are at high risk of strokes. We hypothesized that curative hematopoietic stem cell transplant (HSCT) normalizes cerebral hemodynamics in children with SCD compared with pre-transplant baseline. Whole-brain cerebral blood flow (CBF) and oxygen extraction fraction (OEF) were measured by magnetic resonance imaging 1 to 3 months before and 12 to 24 months after HSCT in 10 children with SCD. Three children had prior overt strokes, 5 children had prior silent strokes, and 1 child had abnormal transcranial Doppler ultrasound velocities. CBF and OEF of HSCT recipients were compared with non-SCD control participants and with SCD participants receiving chronic red blood cell transfusion therapy (CRTT) before and after a scheduled transfusion. Seven participants received matched sibling donor HSCT, and 3 participants received 8 out of 8 matched unrelated donor HSCT. All received reduced-intensity preparation and maintained engraftment, free of hemolytic anemia and SCD symptoms. Pre-transplant, CBF (93.5 mL/100 g/min) and OEF (36.8%) were elevated compared with non-SCD control participants, declining significantly 1 to 2 years after HSCT (CBF, 72.7 mL/100 g per minute; P = .004; OEF, 27.0%; P = .002), with post-HSCT CBF and OEF similar to non-SCD control participants. Furthermore, HSCT recipients demonstrated greater reduction in CBF (-19.4 mL/100 g/min) and OEF (-8.1%) after HSCT than children with SCD receiving CRTT after a scheduled transfusion (CBF, -0.9 mL/100 g/min; P = .024; OEF, -3.3%; P = .001). Curative HSCT normalizes whole-brain hemodynamics in children with SCD. This restoration of cerebral oxygen reserve may explain stroke protection after HSCT in this high-risk patient population.


Asunto(s)
Anemia de Células Falciformes , Trasplante de Células Madre Hematopoyéticas , Accidente Cerebrovascular , Humanos , Niño , Anemia de Células Falciformes/terapia , Accidente Cerebrovascular/prevención & control , Hemodinámica , Oxígeno , Circulación Cerebrovascular
17.
Blood ; 141(2): 194-199, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36315910

RESUMEN

Sickle cell disease (SCD) is an inherited disorder resulting from a ß-globin gene mutation, and SCD patients experience erythrocyte sickling, vaso-occlusive episodes (VOE), and progressive organ damage. Chronic hemolysis, inflammation, and repeated red blood cell transfusions in SCD can disrupt iron homeostasis. Patients who receive multiple blood transfusions develop iron overload, and another subpopulation of SCD patients manifest iron deficiency. To elucidate connections between dietary iron, the microbiome, and SCD pathogenesis, we treated SCD mice with an iron-restricted diet (IRD). IRD treatment reduced iron availability and hemolysis, decreased acute VOE, and ameliorated chronic organ damage in SCD mice. Our results extend previous studies indicating that the gut microbiota regulate disease in SCD mice. IRD alters microbiota load and improves gut integrity, together preventing crosstalk between the gut microbiome and inflammatory factors such as aged neutrophils, dampening VOE, and organ damage. These findings provide strong evidence for the therapeutic potential of manipulating iron homeostasis and the gut microbiome to ameliorate SCD pathophysiology. Many treatments, which are under development, focus on lowering the systemic iron concentration to relieve disease complications, and our data suggest that iron-induced changes in microbiota load and gut integrity are related- and novel-therapeutic targets.


Asunto(s)
Anemia de Células Falciformes , Enfermedades Vasculares , Ratones , Animales , Hierro de la Dieta , Hierro , Hemólisis , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/terapia , Enfermedades Vasculares/etiología , Enfermedades Vasculares/prevención & control
18.
Blood ; 142(3): 230-234, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37216689

RESUMEN

Because of the unique biology of sickle cell disease (SCD) as well as the societal disadvantages and racial inequities suffered by these patients, individuals with SCD have not benefited from the same remarkable advances in care and therapeutics as those with other hematologic disorders. Life expectancy of individuals with SCD is shortened by ∼20 years even with optimal clinical care, and infant mortality continues to be a major concern in low-income countries. As hematologists, we must do more. The American Society of Hematology (ASH) and the ASH Research Collaborative have instituted a multipronged initiative to improve the lives of individuals living with this disease. Here, we describe 2 components of this ASH initiative, the Consortium on Newborn Screening in Africa (CONSA) to improve the early diagnosis of infants in low-resource countries and the SCD Clinical Trial Network to accelerate the development of more effective therapeutics and care for those with this disorder. The combination of SCD-focused initiatives, ASH Research Collaborative, CONSA, and Sickle Cell Clinical Trials Network has enormous potential to dramatically alter the course of SCD worldwide. We believe that the timing is ripe to embark on these critical and worthwhile initiatives and improve the lives of individuals with this disease.


Asunto(s)
Anemia de Células Falciformes , Enfermedades Hematológicas , Lactante , Recién Nacido , Humanos , Anemia de Células Falciformes/terapia , Anemia de Células Falciformes/tratamiento farmacológico , Esperanza de Vida , Atención al Paciente , Tamizaje Neonatal
19.
Blood ; 142(24): 2047-2054, 2023 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-37890140

RESUMEN

As most patients with sickle cell disease (SCD) do not have access to curative therapies, the availability of drug therapies that can modify disease severity remains highly desirable. Despite an increased understanding of the pathophysiology of SCD, only 4 drugs are approved by the US Food and Drugs Administration. Most drug trials in SCD have involved the use of acute pain episodes as the primary clinical end point. These studies have typically been to prevent or shorten the duration of such episodes. To date, no drug has received regulatory approval for shortening the duration of acute vaso-occlusive complications, likely highlighting the complex pathophysiology of acute pain episodes. Trials to prevent acute pain episodes have largely evaluated those episodes requiring health care use as a surrogate end point. However, with differences in culture and health care practices among countries, health care use may not reliably predict clinically important effects on acute pain episodes. This article discusses issues related to the use of health care use as the primary end point for prevention trials of acute pain episodes and highlights the importance of evaluating patient-reported outcomes as well as other SCD-related complications as outcome measures.


Asunto(s)
Dolor Agudo , Anemia de Células Falciformes , Humanos , Dolor Agudo/etiología , Anemia de Células Falciformes/terapia , Anemia de Células Falciformes/tratamiento farmacológico , Evaluación de Resultado en la Atención de Salud
20.
Mol Ther ; 32(5): 1202-1218, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38454604

RESUMEN

In recent years, a growing number of clinical trials have been initiated to evaluate gene therapy approaches for the treatment of patients with transfusion-dependent ß-thalassemia and sickle cell disease (SCD). Therapeutic modalities being assessed in these trials utilize different molecular techniques, including lentiviral vectors to add functional copies of the gene encoding the hemoglobin ß subunit in defective cells and CRISPR-Cas9, transcription activator-like effector protein nuclease, and zinc finger nuclease gene editing strategies to either directly address the underlying genetic cause of disease or induce fetal hemoglobin production by gene disruption. Here, we review the mechanisms of action of these various gene addition and gene editing approaches and describe the status of clinical trials designed to evaluate the potentially for these approaches to provide one-time functional cures to patients with transfusion-dependent ß-thalassemia and SCD.


Asunto(s)
Sistemas CRISPR-Cas , Ensayos Clínicos como Asunto , Edición Génica , Terapia Genética , Vectores Genéticos , Hemoglobinopatías , Humanos , Terapia Genética/métodos , Edición Génica/métodos , Hemoglobinopatías/terapia , Hemoglobinopatías/genética , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Anemia de Células Falciformes/terapia , Anemia de Células Falciformes/genética , Talasemia beta/terapia , Talasemia beta/genética , Animales , Lentivirus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA