Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Nature ; 628(8008): 639-647, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570691

ABSTRACT

Prime editing enables the precise modification of genomes through reverse transcription of template sequences appended to the 3' ends of CRISPR-Cas guide RNAs1. To identify cellular determinants of prime editing, we developed scalable prime editing reporters and performed genome-scale CRISPR-interference screens. From these screens, a single factor emerged as the strongest mediator of prime editing: the small RNA-binding exonuclease protection factor La. Further investigation revealed that La promotes prime editing across approaches (PE2, PE3, PE4 and PE5), edit types (substitutions, insertions and deletions), endogenous loci and cell types but has no consistent effect on genome-editing approaches that rely on standard, unextended guide RNAs. Previous work has shown that La binds polyuridine tracts at the 3' ends of RNA polymerase III transcripts2. We found that La functionally interacts with the 3' ends of polyuridylated prime editing guide RNAs (pegRNAs). Guided by these results, we developed a prime editor protein (PE7) fused to the RNA-binding, N-terminal domain of La. This editor improved prime editing with expressed pegRNAs and engineered pegRNAs (epegRNAs), as well as with synthetic pegRNAs optimized for La binding. Together, our results provide key insights into how prime editing components interact with the cellular environment and suggest general strategies for stabilizing exogenous small RNAs therein.


Subject(s)
Gene Editing , RNA-Binding Proteins , Humans , CRISPR-Cas Systems/genetics , Gene Editing/methods , K562 Cells , Poly U/genetics , Poly U/metabolism , RNA Polymerase III/metabolism , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , RNA-Binding Proteins/metabolism
2.
Cytotherapy ; 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38506770

ABSTRACT

Ex vivo resting culture is a standard procedure following genome editing in hematopoietic stem and progenitor cells (HSPCs). However, prolonged culture may critically affect cell viability and stem cell function. We investigated whether varying durations of culture resting times impact the engraftment efficiency of human CD34+ HSPCs edited at the BCL11A enhancer, a key regulator in the expression of fetal hemoglobin. We employed electroporation to introduce CRISPR-Cas9 components for BCL11A enhancer editing and compared outcomes with nonelectroporated (NEP) and electroporated-only (EP) control groups. Post-electroporation, we monitored cell viability, death rates, and the frequency of enriched hematopoietic stem cell (HSC) fractions (CD34+CD90+CD45RA- cells) over a 48-hour period. Our findings reveal that while the NEP group showed an increase in cell numbers 24 hours post-electroporation, both EP and BCL11A-edited groups experienced significant cell loss. Although CD34+ cell frequency remained high in all groups for up to 48 hours post-electroporation, the frequency of the HSC-enriched fraction was significantly lower in the EP and edited groups compared to the NEP group. In NBSGW xenograft mouse models, both conditioned with busulfan and nonconditioned, we found that immediate transplantation post-electroporation led to enhanced engraftment without compromising editing efficiency. Human glycophorin A+ (GPA+) red blood cells (RBCs) sorted from bone marrow of all BCL11A edited mice exhibited similar levels of γ-globin expression, regardless of infusion time. Our findings underscore the critical importance of optimizing the culture duration between genome editing and transplantation. Minimizing this interval may significantly enhance engraftment success and minimize cell loss without compromising editing efficiency. These insights offer a pathway to improve the success rates of genome editing in HSPCs, particularly for conditions like sickle cell disease.

3.
bioRxiv ; 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38370706

ABSTRACT

Over the last 5 years, cytosine base editors (CBEs) have emerged as a promising therapeutic tool for specific editing of single nucleotide variants and disrupting specific genes associated with disease. Despite this promise, the currently available CBE's have the significant liabilities of off-target and bystander editing activities, in part due to the mechanism by which they are delivered, causing limitations in their potential applications. In this study we engineeredhighly stabilized Cas-embedded CBEs (sCE_CBEs) that integrate several recent advances, andthat are highly expressible and soluble for direct delivery into cells as ribonucleoprotein (RNP) complexes. Our resulting sCE_CBE RNP complexes efficiently and specifically target TC dinucleotides with minimal off-target or bystander mutations. Additional uracil glycosylase inhibitor (UGI) protein in trans further increased C-to-T editing efficiency and target purity in a dose-dependent manner, minimizing indel formation to untreated levels. A single electroporation was sufficient to effectively edit the therapeutically relevant locus for sickle cell disease in hematopoietic stem and progenitor cells (HSPC) in a dose dependent manner without cellular toxicity. Significantly, these sCE_CBE RNPs permitted for the transplantation of edited HSPCs confirming highly efficient editing in engrafting hematopoietic stem cells in mice. The success of the designed sCBE editors, with improved solubility and enhanced on-target editing, demonstrates promising agents for cytosine base editing at other disease-related sites in HSPCs and other cell types.

4.
Nat Methods ; 20(9): 1368-1378, 2023 09.
Article in English | MEDLINE | ID: mdl-37537351

ABSTRACT

Gene regulatory networks (GRNs) are key determinants of cell function and identity and are dynamically rewired during development and disease. Despite decades of advancement, challenges remain in GRN inference, including dynamic rewiring, causal inference, feedback loop modeling and context specificity. To address these challenges, we develop Dictys, a dynamic GRN inference and analysis method that leverages multiomic single-cell assays of chromatin accessibility and gene expression, context-specific transcription factor footprinting, stochastic process network and efficient probabilistic modeling of single-cell RNA-sequencing read counts. Dictys improves GRN reconstruction accuracy and reproducibility and enables the inference and comparative analysis of context-specific and dynamic GRNs across developmental contexts. Dictys' network analyses recover unique insights in human blood and mouse skin development with cell-type-specific and dynamic GRNs. Its dynamic network visualizations enable time-resolved discovery and investigation of developmental driver transcription factors and their regulated targets. Dictys is available as a free, open-source and user-friendly Python package.


Subject(s)
Gene Regulatory Networks , Multiomics , Animals , Mice , Humans , Reproducibility of Results , Transcription Factors/genetics , Algorithms
5.
Nature ; 621(7978): 404-414, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648862

ABSTRACT

Despite the considerable efficacy observed when targeting a dispensable lineage antigen, such as CD19 in B cell acute lymphoblastic leukaemia1,2, the broader applicability of adoptive immunotherapies is hampered by the absence of tumour-restricted antigens3-5. Acute myeloid leukaemia immunotherapies target genes expressed by haematopoietic stem/progenitor cells (HSPCs) or differentiated myeloid cells, resulting in intolerable on-target/off-tumour toxicity. Here we show that epitope engineering of donor HSPCs used for bone marrow transplantation endows haematopoietic lineages with selective resistance to chimeric antigen receptor (CAR) T cells or monoclonal antibodies, without affecting protein function or regulation. This strategy enables the targeting of genes that are essential for leukaemia survival regardless of shared expression on HSPCs, reducing the risk of tumour immune escape. By performing epitope mapping and library screenings, we identified amino acid changes that abrogate the binding of therapeutic monoclonal antibodies targeting FLT3, CD123 and KIT, and optimized a base-editing approach to introduce them into CD34+ HSPCs, which retain long-term engraftment and multilineage differentiation ability. After CAR T cell treatment, we confirmed resistance of epitope-edited haematopoiesis and concomitant eradication of patient-derived acute myeloid leukaemia xenografts. Furthermore, we show that multiplex epitope engineering of HSPCs is feasible and enables more effective immunotherapies against multiple targets without incurring overlapping off-tumour toxicities. We envision that this approach will provide opportunities to treat relapsed/refractory acute myeloid leukaemia and enable safer non-genotoxic conditioning.


Subject(s)
Epitopes , Gene Editing , Immunotherapy , Leukemia, Myeloid, Acute , Animals , Humans , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antigens, CD34/metabolism , Bone Marrow Transplantation , Epitope Mapping , Epitopes/genetics , Epitopes/immunology , Hematopoiesis , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Heterografts/immunology , Immunotherapy/adverse effects , Immunotherapy/methods , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Receptors, Chimeric Antigen/immunology , Recurrence , T-Lymphocytes/immunology , Transplantation Conditioning , Tumor Escape , Xenograft Model Antitumor Assays
6.
Blood ; 142(9): 755-756, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37651157
7.
bioRxiv ; 2023 May 27.
Article in English | MEDLINE | ID: mdl-37292647

ABSTRACT

Gene editing the BCL11A erythroid enhancer is a validated approach to fetal hemoglobin (HbF) induction for ß-hemoglobinopathy therapy, though heterogeneity in edit allele distribution and HbF response may impact its safety and efficacy. Here we compared combined CRISPR-Cas9 endonuclease editing of the BCL11A +58 and +55 enhancers with leading gene modification approaches under clinical investigation. We found that combined targeting of the BCL11A +58 and +55 enhancers with 3xNLS-SpCas9 and two sgRNAs resulted in superior HbF induction, including in engrafting erythroid cells from sickle cell disease (SCD) patient xenografts, attributable to simultaneous disruption of core half E-box/GATA motifs at both enhancers. We corroborated prior observations that double strand breaks (DSBs) could produce unintended on- target outcomes in hematopoietic stem and progenitor cells (HSPCs) such as long deletions and centromere-distal chromosome fragment loss. We show these unintended outcomes are a byproduct of cellular proliferation stimulated by ex vivo culture. Editing HSPCs without cytokine culture bypassed long deletion and micronuclei formation while preserving efficient on-target editing and engraftment function. These results indicate that nuclease editing of quiescent hematopoietic stem cells (HSCs) limits DSB genotoxicity while maintaining therapeutic potency and encourages efforts for in vivo delivery of nucleases to HSCs.

9.
Leukemia ; 37(8): 1698-1708, 2023 08.
Article in English | MEDLINE | ID: mdl-37391485

ABSTRACT

Many inherited bone marrow failure syndromes (IBMFSs) present a high risk of transformation to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). During transformation of IBMFSs, hematopoietic stem and progenitor cells (HSPCs) with poor fitness gain ectopic, dysregulated self-renewal secondary to somatic mutations via undefined mechanisms. Here, in the context of the prototypical IBMFS Fanconi anemia (FA), we performed multiplexed gene editing of mutational hotspots in MDS-associated genes in human induced pluripotent stem cells (iPSCs) followed by hematopoietic differentiation. We observed aberrant self-renewal and impaired differentiation of HSPCs with enrichment of RUNX1 insertions and deletions (indels), generating a model of IBMFS-associated MDS. We observed that compared to the failure state, FA MDS cells show mutant RUNX1-mediated blunting of the G1/S cell cycle checkpoint that is normally activated in FA in response to DNA damage. RUNX1 indels also lead to activation of innate immune signaling, which stabilizes the homologous recombination (HR) effector BRCA1, and this pathway can be targeted to abrogate viability and restore sensitivity to genotoxins in FA MDS. Together, these studies develop a paradigm for modeling clonal evolution in IBMFSs, provide basic understanding of the pathogenesis of MDS, and uncover a therapeutic target in FA-associated MDS.


Subject(s)
Fanconi Anemia , Induced Pluripotent Stem Cells , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia/therapy , Congenital Bone Marrow Failure Syndromes/complications , Core Binding Factor Alpha 2 Subunit/genetics , Induced Pluripotent Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Mutation , Leukemia, Myeloid, Acute/pathology
10.
Mol Ther Methods Clin Dev ; 29: 483-493, 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37273902

ABSTRACT

CRISPR-Cas9-based therapeutic genome editing approaches hold promise to cure a variety of human diseases. Recent findings demonstrate pre-existing immunity for the commonly used Cas orthologs from Streptococcus pyogenes (SpCas9) and Staphylococcus aureus (SaCas9) in humans, which threatens the success of this powerful tool in clinical use. Thus, a comprehensive investigation and potential risk assessment are required to exploit the full potential of the system. Here, we investigated existence of immunity to SpCas9 and SaCas9 in control rhesus macaques (Macaca mulatta) alongside monkeys transplanted with either lentiviral transduced or CRISPR-SpCas9 ribonucleoprotein (RNP)-edited cells. We observed significant levels of Cas9 antibodies in the peripheral blood of all transplanted and non-transplanted control animals. Transplantation of ex vivo transduced or SpCas9-mediated BCL11A enhancer-edited cells did not alter the levels of Cas9 antibodies in rhesus monkeys. Following stimulation of peripheral blood cells with SpCas9 or SaCas9, neither Cas9-specific T cells nor cytokine induction were detected. Robust and durable editing frequencies and expression of high levels of fetal hemoglobin in BCL11A enhancer-edited rhesus monkeys with no evidence of an immune response (>3 years) provide an optimistic outlook for the use of ex vivo CRISPR-SpCas9 (RNP)-edited cells.

12.
Hematol Oncol Clin North Am ; 37(2): 273-299, 2023 04.
Article in English | MEDLINE | ID: mdl-36907603

ABSTRACT

Thalassemia syndromes are common monogenic disorders and represent a significant health issue worldwide. In this review, the authors elaborate on fundamental genetic knowledge about thalassemias, including the structure and location of globin genes, the production of hemoglobin during development, the molecular lesions causing α-, ß-, and other thalassemia syndromes, the genotype-phenotype correlation, and the genetic modifiers of these conditions. In addition, they briefly discuss the molecular techniques applied for diagnosis and innovative cell and gene therapy strategies to cure these conditions.


Subject(s)
Thalassemia , beta-Thalassemia , Humans , beta-Thalassemia/genetics , Syndrome , Thalassemia/genetics , Hemoglobins/genetics , Genetic Association Studies
13.
Nat Commun ; 14(1): 212, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36639728

ABSTRACT

CRISPR-Cas gene editing has revolutionized experimental molecular biology over the past decade and holds great promise for the treatment of human genetic diseases. Here we review the development of CRISPR-Cas9/Cas12/Cas13 nucleases, DNA base editors, prime editors, and RNA base editors, focusing on the assessment and improvement of their editing precision and safety, pushing the limit of editing specificity and efficiency. We summarize the capabilities and limitations of each CRISPR tool from DNA editing to RNA editing, and highlight the opportunities for future improvements and applications in basic research, as well as the therapeutic and clinical considerations for their use in patients.


Subject(s)
CRISPR-Cas Systems , RNA Editing , Humans , CRISPR-Cas Systems/genetics , RNA Editing/genetics , Gene Editing , DNA/genetics , Endonucleases/metabolism
14.
Nat Commun ; 14(1): 207, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36639729

ABSTRACT

In ß-thalassemia, either γ-globin induction to form fetal hemoglobin (α2γ2) or ß-globin repair to restore adult hemoglobin (α2ß2) could be therapeutic. ABE8e, a recently evolved adenine base editor variant, can achieve efficient adenine conversion, yet its application in patient-derived hematopoietic stem cells needs further exploration. Here, we purified ABE8e for ribonucleoprotein electroporation of ß-thalassemia patient CD34+ hematopoietic stem and progenitor cells to introduce nucleotide substitutions that upregulate γ-globin expression in the BCL11A enhancer or in the HBG promoter. We observed highly efficient on-target adenine base edits at these two regulatory regions, resulting in robust γ-globin induction. Moreover, we developed ABE8e-SpRY, a near-PAMless ABE variant, and successfully applied ABE8e-SpRY RNP to directly correct HbE and IVS II-654 mutations in patient-derived CD34+ HSPCs. Finally, durable therapeutic editing was produced in self-renewing repopulating human HSCs as assayed in primary and secondary recipients. Together, these results support the potential of ABE-mediated base editing in HSCs to treat inherited monogenic blood disorders.


Subject(s)
Gene Editing , beta-Thalassemia , Humans , Adenine/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , beta-Thalassemia/genetics , CRISPR-Cas Systems/genetics , Fetal Hemoglobin/genetics , gamma-Globins/genetics , Gene Editing/methods , Hematopoietic Stem Cells/metabolism
15.
Methods Mol Biol ; 2606: 43-62, 2023.
Article in English | MEDLINE | ID: mdl-36592307

ABSTRACT

Base editing by nucleotide deaminases linked to programmable DNA-binding proteins represents a promising approach to remedy blood disorders. Here we describe the ex vivo base editing of human CD34+ hematopoietic stem and progenitor cells (HSPCs) by electroporation of base editor mRNA or protein.


Subject(s)
Gene Editing , Hematopoietic Stem Cell Transplantation , Humans , Hematopoietic Stem Cells/metabolism , Antigens, CD34/metabolism
16.
Nat Genet ; 55(1): 34-43, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36522432

ABSTRACT

CRISPR gene editing holds great promise to modify DNA sequences in somatic cells to treat disease. However, standard computational and biochemical methods to predict off-target potential focus on reference genomes. We developed an efficient tool called CRISPRme that considers single-nucleotide polymorphism (SNP) and indel genetic variants to nominate and prioritize off-target sites. We tested the software with a BCL11A enhancer targeting guide RNA (gRNA) showing promise in clinical trials for sickle cell disease and ß-thalassemia and found that the top candidate off-target is produced by an allele common in African-ancestry populations (MAF 4.5%) that introduces a protospacer adjacent motif (PAM) sequence. We validated that SpCas9 generates strictly allele-specific indels and pericentric inversions in CD34+ hematopoietic stem and progenitor cells (HSPCs), although high-fidelity Cas9 mitigates this off-target. This report illustrates how genetic variants should be considered as modifiers of gene editing outcomes. We expect that variant-aware off-target assessment will become integral to therapeutic genome editing evaluation and provide a powerful approach for comprehensive off-target nomination.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Humans , Gene Editing/methods , CRISPR-Cas Systems/genetics , Hematopoietic Stem Cells , INDEL Mutation , RNA, Guide, CRISPR-Cas Systems
17.
Cell Stem Cell ; 29(10): 1426-1427, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36206729

ABSTRACT

Therapeutic gene editing based on homology-directed repair (HDR) promises to precisely recombine donor template sequences at programmed double-strand break sites. In this issue, Ferrari et al. report that commonly used AAV donors blunt hematopoietic repopulation and result in frequent genomic integrations that could interfere with normal gene expression.


Subject(s)
DNA Breaks, Double-Stranded , Gene Editing , CRISPR-Cas Systems , DNA , DNA End-Joining Repair , Hematopoietic Stem Cells , Recombinational DNA Repair
18.
Mol Ther ; 30(8): 2693-2708, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35526095

ABSTRACT

A promising treatment for ß-hemoglobinopathies is the de-repression of γ-globin expression leading to increased fetal hemoglobin (HbF) by targeting BCL11A. Here, we aim to improve a lentivirus vector (LV) containing a single BCL11A shmiR (SS) to further increase γ-globin induction. We engineered a novel LV to express two shmiRs simultaneously targeting BCL11A and the γ-globin repressor ZNF410. Erythroid cells derived from human HSCs transduced with the double shmiR (DS) showed up to a 70% reduction of both BCL11A and ZNF410 proteins. There was a consistent and significant additional 10% increase in HbF compared to targeting BCL11A alone in erythroid cells. Erythrocytes differentiated from SCD HSCs transduced with the DS demonstrated significantly reduced in vitro sickling phenotype compared to the SS. Erythrocytes differentiated from transduced HSCs from ß-thalassemia major patients demonstrated improved globin chain balance by increased γ-globin with reduced microcytosis. Reconstitution of DS-transduced cells from Berkeley SCD mice was associated with a statistically larger reduction in peripheral blood hemolysis markers compared with the SS vector. Overall, these results indicate that the DS LV targeting BCL11A and ZNF410 can enhance HbF induction for treating ß-hemoglobinopathies and could be used as a model to simultaneously and efficiently target multiple gene products.


Subject(s)
Fetal Hemoglobin , Hemoglobinopathies , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/metabolism , gamma-Globins/genetics
19.
PLoS One ; 17(2): e0263829, 2022.
Article in English | MEDLINE | ID: mdl-35167623

ABSTRACT

Fibrolamellar carcinoma (FLC) is a primary liver cancer that most commonly arises in adolescents and young adults in a background of normal liver tissue and has a poor prognosis due to lack of effective chemotherapeutic agents. The DNAJB1-PRKACA gene fusion (DP) has been reported in the majority of FLC tumors; however, its oncogenic mechanisms remain unclear. Given the paucity of cellular models, in particular FLC tumor cell lines, we hypothesized that engineering the DP fusion gene in HEK293T cells would provide insight into the cellular effects of the fusion gene. We used CRISPR/Cas9 to engineer HEK293T clones expressing DP fusion gene (HEK-DP) and performed transcriptomic, proteomic, and mitochondrial studies to characterize this cellular model. Proteomic analysis of DP interacting partners identified mitochondrial proteins as well as proteins in other subcellular compartments. HEK-DP cells demonstrated significantly elevated mitochondrial fission, which suggests a role for DP in altering mitochondrial dynamics. Transcriptomic analysis of HEK-DP cells revealed a significant increase in LINC00473 expression, similar to what has been observed in primary FLC samples. LINC00473 overexpression was reversible with siRNA targeting of PRKACA as well as pharmacologic targeting of PKA and Hsp40 in HEK-DP cells. Therefore, our model suggests that LINC00473 is a candidate marker for DP activity.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , HSP40 Heat-Shock Proteins/genetics , Oncogene Proteins, Fusion/genetics , RNA, Long Noncoding/genetics , Up-Regulation , CRISPR-Cas Systems , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , HSP40 Heat-Shock Proteins/metabolism , Humans , Mitochondria/metabolism , Models, Biological , Proteomics
20.
Leukemia ; 36(2): 383-393, 2022 02.
Article in English | MEDLINE | ID: mdl-34344987

ABSTRACT

Acute myeloid leukemia (AML) is a devastating disease, and clinical outcomes are still far from satisfactory. Here, to identify novel targets for AML therapy, we performed a genome-wide CRISPR/Cas9 screen using AML cell lines, followed by a second screen in vivo. We show that PAICS, an enzyme involved in de novo purine biosynthesis, is a potential target for AML therapy. AML cells expressing shRNA-PAICS exhibited a proliferative disadvantage, indicating a toxic effect of shRNA-PAICS. Treatment of human AML cells with a PAICS inhibitor suppressed their proliferation by inhibiting DNA synthesis and promoting apoptosis and had anti-leukemic effects in AML PDX models. Furthermore, CRISPR/Cas9 screens using AML cells in the presence of the inhibitor revealed genes mediating resistance or synthetic lethal to PAICS inhibition. Our findings identify PAICS as a novel therapeutic target for AML and further define components of de novo purine synthesis pathway and its downstream effectors essential for AML cell survival.


Subject(s)
CRISPR-Cas Systems , Carboxy-Lyases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Purines/metabolism , Animals , Apoptosis , Cell Proliferation , Genome-Wide Association Study , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...