Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Med ; 29(12): 3175-3183, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37973947

ABSTRACT

Gene therapy (GT) provides a potentially curative treatment option for patients with sickle cell disease (SCD); however, the occurrence of myeloid malignancies in GT clinical trials has prompted concern, with several postulated mechanisms. Here, we used whole-genome sequencing to track hematopoietic stem cells (HSCs) from six patients with SCD at pre- and post-GT time points to map the somatic mutation and clonal landscape of gene-modified and unmodified HSCs. Pre-GT, phylogenetic trees were highly polyclonal and mutation burdens per cell were elevated in some, but not all, patients. Post-GT, no clonal expansions were identified among gene-modified or unmodified cells; however, an increased frequency of potential driver mutations associated with myeloid neoplasms or clonal hematopoiesis (DNMT3A- and EZH2-mutated clones in particular) was observed in both genetically modified and unmodified cells, suggesting positive selection of mutant clones during GT. This work sheds light on HSC clonal dynamics and the mutational landscape after GT in SCD, highlighting the enhanced fitness of some HSCs harboring pre-existing driver mutations. Future studies should define the long-term fate of mutant clones, including any contribution to expansions associated with myeloid neoplasms.


Subject(s)
Anemia, Sickle Cell , Neoplasms , Humans , Hematopoiesis/genetics , Phylogeny , Mutation/genetics , Hematopoietic Stem Cells/pathology , Clone Cells , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Anemia, Sickle Cell/pathology , Genetic Therapy , Neoplasms/pathology
2.
Nat Commun ; 14(1): 5850, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37730674

ABSTRACT

We previously reported initial clinical results of post-transcriptional gene silencing of BCL11A expression (NCT03282656) reversing the fetal to adult hemoglobin switch. A goal of this approach is to increase fetal hemoglobin (HbF) expression while coordinately reducing sickle hemoglobin (HbS) expression. The resulting combinatorial effect should prove effective in inhibiting HbS polymerization at lower physiologic oxygen values thereby mitigating disease complications. Here we report results of exploratory single-cell analysis of patients in which BCL11A is targeted molecularly and compare results with cells of patients treated with hydroxyurea (HU), the current standard of care. We use single-cell assays to assess HbF, HbS, oxygen saturation, and hemoglobin polymer content in RBCs for nine gene therapy trial subjects (BCLshmiR, median HbF% = 27.9) and compare them to 10 HU-treated subjects demonstrating high and comparable levels of HbF (HU High Responders, median HbF% = 27.0). All BCL11A patients achieved the primary endpoint for NCT03282656, which was defined by an absolute neutrophil count greater than or equal to 0.5 × 109 cells/L for three consecutive days, achieved within 7 weeks following infusion. Flow cytometric assessment of single-RBC HbF and HbS shows fewer RBCs with high HbS% that would be most susceptible to sickling in BCLshmiR vs. HU High Responders: median 42% of RBCs with HbS%>70% in BCLshmiR vs. 61% in HU High Responders (p = 0.004). BCLshmiR subjects also demonstrate more RBCs resistant to HbS polymerization at lower physiologic oxygen tension: median 32% vs. 25% in HU High Responders (p = 0.006). Gene therapy-induced BCL11A down-regulation reverses the fetal-to-adult hemoglobin switch and induces RBCs with higher HbF%, lower HbS%, and greater resistance to deoxygenation-induced polymerization in clinical trial subjects compared with a cohort of highly responsive hydroxyurea-treated subjects.


Subject(s)
Hemoglobin, Sickle , Hydroxyurea , Adult , Humans , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , Erythrocytes , Fetus , Fetal Hemoglobin/genetics , Transcription Factors
4.
Pediatr Blood Cancer ; 70(10): e30539, 2023 10.
Article in English | MEDLINE | ID: mdl-37470711

ABSTRACT

BACKGROUND: Children with sickle cell anemia (SCA) have substantial medical needs and more unmet basic needs than children with other medical conditions. Despite a recent focus on social determinants of health (SDoH), there remains an incomplete understanding of the processes linking SDoH and disease management, particularly for youth with SCA. This study elucidated these processes and identified ways to mitigate deleterious effects of adverse SDoH on SCA management. METHODS: Parents/primary caregivers (N = 27) of children with SCA (≤12 years old) participated in semi-structured interviews regarding SCA management and SDoH and completed quantitative measures of basic needs. Qualitative data were systematically coded and analyzed using applied thematic analysis. Quantitative data were presented descriptively. RESULTS: Three qualitative themes were identified. First, SCA management is bidirectionally linked with the social environment, whereby challenges of SCA management can hinder basic needs from being met, and unmet basic needs and financial hardship hinder SCA management. Second, due to limited resources, parents/caregivers are faced with difficult choices between prioritizing basic needs versus SCA management. Third, addressing material, emotional, and informational needs may improve SCA management. Quantitatively, 73% of families endorsed ≥1 basic need, including food insecurity (42%), housing instability (62%), and/or energy insecurity 19% (vs. 20%). CONCLUSION: Despite documented associations, there remains a poor understanding of the processes linking SDoH and health. Findings underscore how day-to-day conditions undermine the management of SCA treatments, symptoms, and complications, limiting treatment effectiveness. Understanding these processes may inform family-centered, health equity interventions and policies to improve living conditions, disease management, and health outcomes.


Subject(s)
Anemia, Sickle Cell , Social Determinants of Health , Adolescent , Child , Humans , Parents , Qualitative Research , Anemia, Sickle Cell/therapy , Surveys and Questionnaires
5.
J Clin Oncol ; 41(12): 2227-2237, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36623245

ABSTRACT

PURPOSE: To report the incidence and risk factors for secondary neoplasm after transplantation for sickle cell disease. METHODS: Included are 1,096 transplants for sickle cell disease between 1991 and 2016. There were 22 secondary neoplasms. Types included leukemia/myelodysplastic syndrome (MDS; n = 15) and solid tumor (n = 7). Fine-Gray regression models examined for risk factors for leukemia/MDS and any secondary neoplasm. RESULTS: The 10-year incidence of leukemia/MDS was 1.7% (95% CI, 0.90 to 2.9) and of any secondary neoplasm was 2.4% (95% CI, 1.4 to 3.8). After adjusting for other risk factors, risks for leukemia/MDS (hazard ratio, 22.69; 95% CI, 4.34 to 118.66; P = .0002) or any secondary neoplasm (hazard ratio, 7.78; 95% CI, 2.20 to 27.53; P = .0015) were higher with low-intensity (nonmyeloablative) regimens compared with more intense regimens. All low-intensity regimens included total-body irradiation (TBI 300 or 400 cGy with alemtuzumab, TBI 300 or 400 cGy with cyclophosphamide, TBI 200, 300, or 400 cGy with cyclophosphamide and fludarabine, or TBI 200 cGy with fludarabine). None of the patients receiving myeloablative and only 23% of those receiving reduced-intensity regimens received TBI. CONCLUSION: Low-intensity regimens rely on tolerance induction and establishment of mixed-donor chimerism. Persistence of host cells exposed to low-dose radiation triggering myeloid malignancy is one plausible etiology. Pre-existing myeloid mutations and prior inflammation may also contribute but could not be studied using our data source. Choosing conditioning regimens likely to result in full-donor chimerism may in part mitigate the higher risk for leukemia/MDS.


Subject(s)
Anemia, Sickle Cell , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Neoplasms, Second Primary , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Graft vs Host Disease/etiology , Cyclophosphamide , Anemia, Sickle Cell/etiology , Neoplasms, Second Primary/epidemiology , Neoplasms, Second Primary/etiology , Transplantation Conditioning/adverse effects , Whole-Body Irradiation
6.
Blood Adv ; 6(12): 3803-3811, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35500223

ABSTRACT

Fanconi anemia (FA), a genetic DNA repair disorder characterized by marrow failure and cancer susceptibility. In FA mice, metformin improves blood counts and delays tumor development. We conducted a single institution study of metformin in nondiabetic patients with FA to determine feasibility and tolerability of metformin treatment and to assess for improvement in blood counts. Fourteen of 15 patients with at least 1 cytopenia (hemoglobin < 10 g/dL; platelet count < 100 000 cells/µL; or an absolute neutrophil count < 1000 cells/µL) were eligible to receive metformin for 6 months. Median patient age was 9.4 years (range 6.0-26.5 ). Thirteen of 14 subjects (93%) tolerated maximal dosing for age; 1 subject had dose reduction for grade 2 gastrointestinal symptoms. No subjects developed hypoglycemia or metabolic acidosis. No subjects had dose interruptions caused by toxicity, and no grade 3 or higher adverse events attributed to metformin were observed. Hematologic response based on modified Myelodysplastic Syndrome International Working Group criteria was observed in 4 of 13 evaluable patients (30.8%; 90% confidence interval, 11.3-57.3). Median time to response was 84.5 days (range 71-128 days). Responses were noted in neutrophils (n = 3), platelets (n = 1), and red blood cells (n = 1). No subjects met criteria for disease progression or relapse during treatment. Correlative studies explored potential mechanisms of metformin activity in FA. Plasma proteomics showed reduction in inflammatory pathways with metformin. Metformin is safe and tolerable in nondiabetic patients with FA and may provide therapeutic benefit. This trial was registered at as #NCT03398824.


Subject(s)
Fanconi Anemia , Metformin , Child , Fanconi Anemia/drug therapy , Fanconi Anemia/genetics , Humans , Metformin/therapeutic use , Young Adult
7.
Hematology Am Soc Hematol Educ Program ; 2021(1): 600-606, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34889443

ABSTRACT

After years of reliance on transfusion alone to address anemia and suppress ineffective erythropoiesis in ß-thalassemia, many new therapies are now in development. Luspatercept, a transforming growth factor-ß inhibitor, has demonstrated efficacy in reducing ineffective erythropoiesis, improving anemia, and possibly reducing iron loading. However, many patients do not respond to luspatercept, so additional therapeutics are needed. Several medications in development aim to induce hemoglobin F (HbF): sirolimus, benserazide, and IMR-687 (a phosphodiesterase 9 inhibitor). Another group of agents seeks to ameliorate ineffective erythropoiesis and improve anemia by targeting abnormal iron metabolism in thalassemia: apotransferrin, VIT-2763 (a ferroportin inhibitor), PTG-300 (a hepcidin mimetic), and an erythroferrone antibody in early development. Mitapivat, a pyruvate kinase activator, represents a unique mechanism to mitigate ineffective erythropoiesis. Genetically modified autologous hematopoietic stem cell transplantation offers the potential for lifelong transfusion independence. Through a gene addition approach, lentiviral vectors have been used to introduce a ß-globin gene into autologous hematopoietic stem cells. One such product, betibeglogene autotemcel (beti-cel), has reached phase 3 trials with promising results. In addition, 2 gene editing techniques (CRISPR-Cas9 and zinc-finger nucleases) are under investigation as a means to silence BCL11A to induce HbF with agents designated CTX001 and ST-400, respectively. Results from the many clinical trials for these agents will yield results in the next few years, which may end the era of relying on transfusion alone as the mainstay of thalassemia therapy.


Subject(s)
beta-Thalassemia/therapy , Activin Receptors, Type II/therapeutic use , Adolescent , Adult , Blood Transfusion , Female , Hematinics/therapeutic use , Hematopoietic Stem Cell Transplantation , Humans , Immunoglobulin Fc Fragments/therapeutic use , Iron Chelating Agents/therapeutic use , Male , Middle Aged , Recombinant Fusion Proteins/therapeutic use , Young Adult
8.
Blood Adv ; 5(23): 5452, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34905048

ABSTRACT

Sickle cell disease (SCD) is an inherited blood condition resulting from abnormal hemoglobin production. It is one of the most common genetic diseases in the world. The clinical manifestations are variable and range from recurrent acute and debilitating painful crises to life-threatening pulmonary, cardiovascular, renal, and neurologic complications. The only curative treatment of SCD at this time is bone marrow transplantation (also called hematopoietic stem cell transplantation) using healthy blood stem cells from an unaffected brother or sister or from an unrelated donor if one can be identified who is a match in tissue typing. Unfortunately, only a minority of patients with sickle cell has such a donor available. The use of autologous hematopoietic stem cells and alternative types of genetic modifications is currently under study in clinical research trials for this disease. The approaches include the use of viral vectors to express globin genes that are modified to prevent sickle hemoglobin polymerization or to express interfering RNAs to "flip the switch" in adult red cells from adult ß-sickle hemoglobin to fetal hemoglobin using a physiologic switch, and several gene editing approaches with the goal of inducing fetal hemoglobin or correcting/modifying the actual sickle mutation. In this audio review, we will discuss these different approaches and review the current progress of curative therapy for SCD using gene therapy.


Subject(s)
Anemia, Sickle Cell , Genetic Therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Fetal Hemoglobin , Hemoglobin, Sickle/genetics , Humans , Unrelated Donors
9.
N Engl J Med ; 384(3): 205-215, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33283990

ABSTRACT

BACKGROUND: Sickle cell disease is characterized by hemolytic anemia, pain, and progressive organ damage. A high level of erythrocyte fetal hemoglobin (HbF) comprising α- and γ-globins may ameliorate these manifestations by mitigating sickle hemoglobin polymerization and erythrocyte sickling. BCL11A is a repressor of γ-globin expression and HbF production in adult erythrocytes. Its down-regulation is a promising therapeutic strategy for induction of HbF. METHODS: We enrolled patients with sickle cell disease in a single-center, open-label pilot study. The investigational therapy involved infusion of autologous CD34+ cells transduced with the BCH-BB694 lentiviral vector, which encodes a short hairpin RNA (shRNA) targeting BCL11A mRNA embedded in a microRNA (shmiR), allowing erythroid lineage-specific knockdown. Patients were assessed for primary end points of engraftment and safety and for hematologic and clinical responses to treatment. RESULTS: As of October 2020, six patients had been followed for at least 6 months after receiving BCH-BB694 gene therapy; median follow-up was 18 months (range, 7 to 29). All patients had engraftment, and adverse events were consistent with effects of the preparative chemotherapy. All the patients who could be fully evaluated achieved robust and stable HbF induction (percentage HbF/(F+S) at most recent follow-up, 20.4 to 41.3%), with HbF broadly distributed in red cells (F-cells 58.9 to 93.6% of untransfused red cells) and HbF per F-cell of 9.0 to 18.6 pg per cell. Clinical manifestations of sickle cell disease were reduced or absent during the follow-up period. CONCLUSIONS: This study validates BCL11A inhibition as an effective target for HbF induction and provides preliminary evidence that shmiR-based gene knockdown offers a favorable risk-benefit profile in sickle cell disease. (Funded by the National Institutes of Health; ClinicalTrials.gov number, NCT03282656).


Subject(s)
Anemia, Sickle Cell/therapy , Fetal Hemoglobin/biosynthesis , Genetic Therapy , RNA Interference , Repressor Proteins/genetics , gamma-Globins/metabolism , Adolescent , Adult , Anemia, Sickle Cell/genetics , Child , Down-Regulation , Female , Fetal Hemoglobin/genetics , Gene Knockdown Techniques , Genetic Vectors , Humans , Male , Pilot Projects , RNA, Small Interfering , Repressor Proteins/metabolism , Transplantation, Autologous , Young Adult , gamma-Globins/genetics
10.
Mol Ther Methods Clin Dev ; 17: 589-600, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32300607

ABSTRACT

In this work we provide preclinical data to support initiation of a first-in-human trial for sickle cell disease (SCD) using an approach that relies on reversal of the developmental fetal-to-adult hemoglobin switch. Erythroid-specific knockdown of BCL11A via a lentiviral-encoded microRNA-adapted short hairpin RNA (shRNAmiR) leads to reactivation of the gamma-globin gene while simultaneously reducing expression of the pathogenic adult sickle ß-globin. We generated a refined lentiviral vector (LVV) BCH-BB694 that was developed to overcome poor vector titers observed in the manufacturing scale-up of the original research-grade LVV. Healthy or sickle cell donor CD34+ cells transduced with Good Manufacturing Practices (GMP)-grade BCH-BB694 LVV achieved high vector copy numbers (VCNs) >5 and gene marking of >80%, resulting in a 3- to 5-fold induction of fetal hemoglobin (HbF) compared with mock-transduced cells without affecting growth, differentiation, and engraftment of gene-modified cells in vitro or in vivo. In vitro immortalization assays, which are designed to measure vector-mediated genotoxicity, showed no increased immortalization compared with mock-transduced cells. Together these data demonstrate that BCH-BB694 LVV is non-toxic and efficacious in preclinical studies, and can be generated at a clinically relevant scale in a GMP setting at high titer to support clinical testing for the treatment of SCD.

11.
Nat Med ; 25(5): 776-783, 2019 05.
Article in English | MEDLINE | ID: mdl-30911135

ABSTRACT

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


Subject(s)
Gene Editing/methods , Hematopoietic Stem Cells/metabolism , Amino Acid Sequence , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Base Sequence , CRISPR-Cas Systems , Carrier Proteins/genetics , Enhancer Elements, Genetic , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Hematopoietic Stem Cell Transplantation , Humans , INDEL Mutation , Nuclear Proteins/genetics , RNA, Guide, Kinetoplastida/genetics , Repressor Proteins , beta-Thalassemia/blood , beta-Thalassemia/genetics , beta-Thalassemia/therapy , gamma-Globins/biosynthesis , gamma-Globins/genetics
12.
Blood Adv ; 2(19): 2505-2512, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30282642

ABSTRACT

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


Subject(s)
Anemia, Sickle Cell/therapy , Blood Component Removal , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/administration & dosage , Adolescent , Adult , Benzylamines , Cyclams , Dose-Response Relationship, Drug , Genetic Therapy/methods , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Peripheral Blood Stem Cell Transplantation/methods , Pilot Projects , Young Adult
13.
Semin Hematol ; 55(2): 76-86, 2018 04.
Article in English | MEDLINE | ID: mdl-29958563

ABSTRACT

After decades with few novel therapeutic options for sickle cell disease (SCD), autologous hematopoietic stem cell (HSC) based genetic therapies including lentiviral gene therapy (GT), and genome editing (GE) now appear imminent. Lentiviral GT has advanced considerably in the past decade with promising clinical trial results in multiple disorders. For ß-hemoglobinopathies, GT strategies of gene addition and fetal hemoglobin induction through BCL11A regulation are both being evaluated in open clinical trials. GE techniques offer the possibility of a nonviral curative approach, either through sickle hemoglobin mutation repair or fetal hemoglobin elevation. Although GE currently remains at the preclinical stage, multiple clinical trials will likely open soon. In addition to reviewing current strategies for GT and GE, this review highlights important next steps toward optimization of these therapies. All autologous cell-based genetic therapies rely on safely obtaining an adequate yield of autologous HSCs for genetic modification and transplantation. HSC collection is uniquely challenging in SCD. Peripheral mobilization with plerixafor has recently emerged as a promising approach. The acute and long-term toxicities associated with myeloablative conditioning are risks that may not be acceptable to a significant number of SCD patients, highlighting the need for novel conditioning regimens. Finally, increasing availability of autologous genetic therapies will require comprehensive and collaborative discussions regarding cost and access for SCD patients, at individual centers and worldwide.


Subject(s)
Anemia, Sickle Cell/therapy , Genetic Therapy/methods , Anemia, Sickle Cell/pathology , Humans
15.
Blood ; 125(17): 2729, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-26090541
16.
Am J Hematol ; 90(7): 624-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808664

ABSTRACT

Other than hydroxyurea, no pharmacologic agents are clinically available for fetal hemoglobin (HbF) induction in sickle cell disease (SCD). An optimal candidate would induce HbF without causing cell cycle inhibition and would act independently of hydroxyurea in order to yield additional HbF induction when combined. We explored whether inhibition of histone deacetylase (HDAC) 1 or HDAC2 could achieve these goals. In human erythroid progenitor cells, shRNA knockdown of the HDAC1 or HDAC2 genes induced gamma globin, without altering cellular proliferation in vitro, and without altering cell cycle phase. Treatment with hydroxyurea in combination with HDAC2 knockdown yielded a further increase in gamma globin expression. Additionally, when CD34+ cells were treated with both hydroxyurea and MS-275 (an inhibitor of HDAC 1, 2, and 3), an additive induction of relative gamma globin expression was achieved. Our findings support further clinical investigation of HDAC inhibitors in combination with hydroxyurea in SCD patients.


Subject(s)
Bone Marrow Cells/metabolism , Cell Cycle/genetics , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , gamma-Globins/agonists , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Cycle/drug effects , Cell Differentiation , Gene Expression/drug effects , Gene Knockdown Techniques , Genetic Vectors , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Humans , Hydroxyurea/pharmacology , Lentivirus/genetics , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , beta-Globins/genetics , beta-Globins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism
17.
Science ; 334(6058): 993-6, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21998251

ABSTRACT

Persistence of human fetal hemoglobin (HbF, α(2)γ(2)) in adults lessens the severity of sickle cell disease (SCD) and the ß-thalassemias. Here, we show that the repressor BCL11A is required in vivo for silencing of γ-globin expression in adult animals, yet dispensable for red cell production. BCL11A serves as a barrier to HbF reactivation by known HbF inducing agents. In a proof-of-principle test of BCL11A as a potential therapeutic target, we demonstrate that inactivation of BCL11A in SCD transgenic mice corrects the hematologic and pathologic defects associated with SCD through high-level pancellular HbF induction. Thus, interference with HbF silencing by manipulation of a single target protein is sufficient to reverse SCD.


Subject(s)
Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Carrier Proteins/physiology , Fetal Hemoglobin/genetics , Gene Expression Regulation , Gene Silencing , Nuclear Proteins/physiology , gamma-Globins/genetics , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/pathology , Animals , Carrier Proteins/genetics , DNA Methylation , DNA-Binding Proteins , Embryo, Mammalian , Epigenesis, Genetic , Erythroid Cells/metabolism , Fetal Hemoglobin/metabolism , Histones/metabolism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Molecular Targeted Therapy , Nuclear Proteins/genetics , Repressor Proteins
18.
Ophthalmic Surg Lasers Imaging ; 37(6): 462-7, 2006.
Article in English | MEDLINE | ID: mdl-17152539

ABSTRACT

BACKGROUND AND OBJECTIVE: To determine the visual outcome of laser treatments for macular edema due to branch retinal vein occlusion (BRVO) in patients with a preoperative visual acuity (VA) of 20/200 or worse compared to patients with a preoperative VA of better than 20/200. PATIENTS AND METHODS: Records of 88 patients with macular edema secondary to BRVO undergoing laser treatment from 1984 to 2003 were reviewed. Mean VA was measured before and after each treatment and after the final treatment. RESULTS: All patients received between one and five laser treatments. Preoperative VA was better than 20/200 in 56 patients (group 1) and 20/200 or worse in 32 patients (group 2). Patients in group 1 had a mean improvement of 0.48 lines and 57% had a final VA of 20/40 or better. Patients in group 2 had a mean improvement of 1.69 lines and 20% had a final VA of 20/40 or better. CONCLUSIONS: Patients with poor VA (20/200 or worse) secondary to macular edema due to BRVO responded positively to laser treatment. The level of preoperative VA can be a useful predictor of visual outcome. These patients should consider laser treatment before alternative, more aggressive approaches.


Subject(s)
Laser Therapy/methods , Macular Edema/physiopathology , Retinal Vein Occlusion/complications , Visual Acuity/physiology , Aged , Female , Fluorescein Angiography , Follow-Up Studies , Fundus Oculi , Humans , Macular Edema/etiology , Macular Edema/surgery , Male , Prognosis , Retinal Vein Occlusion/physiopathology , Retinal Vein Occlusion/surgery , Retrospective Studies , Severity of Illness Index
19.
Am J Ophthalmol ; 139(4): 653-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15808160

ABSTRACT

PURPOSE: To report the visual outcome of multiple laser treatments for macular edema attributable to branch retinal vein occlusions (BRVO) and to determine if any prognostic factors exist for improvement. DESIGN: Retrospective chart review. METHODS: A private practice with four vitreoretinal surgeons performed laser treatments on 88 eyes of 88 patients with macular edema secondary to BRVO from 1984 to 2003. Mean preoperative and postoperative visual acuities were collected after each laser treatment. Final visual acuities were also documented. RESULTS: All 88 patients received at least one laser treatment, and 46 patients of the initial 88 underwent multiple treatments. Overall, forty-one (46.6%) of the total 88 patients improved by 2 or more lines, whereas 33 patients (37.5%) were within 1 line of the preoperative vision, and 14 patients (15.9%) worsened by 2 or more lines. The mean final visual improvement was 0.92 lines for all 88 patients. The group of patients that responded favorably to the first laser treatment (n = 37) showed an overall improvement of 3.5 lines. However, patients who responded poorly to the first laser treatment resulted in an overall worsening of vision by 0.96 lines. CONCLUSIONS: Our study found that multiple laser treatments can improve visual acuity and resolve macular edema and that each additional laser treatment gives a patient a modest chance of visual improvement. A positive or stable visual response to first laser treatment portends a favorable prognosis with subsequent laser treatments.


Subject(s)
Laser Coagulation/methods , Macular Edema/surgery , Retinal Vein Occlusion/surgery , Aged , Female , Humans , Macular Edema/etiology , Macular Edema/physiopathology , Male , Reoperation , Retinal Vein Occlusion/complications , Retinal Vein Occlusion/physiopathology , Retrospective Studies , Treatment Outcome , Visual Acuity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...