Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Nat Biomed Eng ; 2023 Dec 13.
Article En | MEDLINE | ID: mdl-38092857

The reliance on viral vectors for the production of genetically engineered immune cells for adoptive cellular therapies remains a translational bottleneck. Here we report a method leveraging the DNA repair pathway homology-mediated end joining, as well as optimized reagent composition and delivery, for the Cas9-induced targeted integration of large DNA payloads into primary human T cells with low toxicity and at efficiencies nearing those of viral vectors (targeted knock-in of 1-6.7 kb payloads at rates of up to 70% at multiple targeted genomic loci and with cell viabilities of over 80%). We used the method to produce T cells with an engineered T-cell receptor or a chimaeric antigen receptor and show that the cells maintained low levels of exhaustion markers and excellent capacities for proliferation and cytokine production and that they elicited potent antitumour cytotoxicity in vitro and in mice. The method is readily adaptable to current good manufacturing practices and scale-up processes, and hence may be used as an alternative to viral vectors for the production of genetically engineered T cells for cancer immunotherapies.

2.
Med ; 3(10): 682-704.e8, 2022 10 14.
Article En | MEDLINE | ID: mdl-36007524

BACKGROUND: Adoptive transfer of tumor-infiltrating lymphocytes (TIL) fails to consistently elicit tumor rejection. Manipulation of intrinsic factors that inhibit T cell effector function and neoantigen recognition may therefore improve TIL therapy outcomes. We previously identified the cytokine-induced SH2 protein (CISH) as a key regulator of T cell functional avidity in mice. Here, we investigate the mechanistic role of CISH in regulating human T cell effector function in solid tumors and demonstrate that CRISPR/Cas9 disruption of CISH enhances TIL neoantigen recognition and response to checkpoint blockade. METHODS: Single-cell gene expression profiling was used to identify a negative correlation between high CISH expression and TIL activation in patient-derived TIL. A GMP-compliant CRISPR/Cas9 gene editing process was developed to assess the impact of CISH disruption on the molecular and functional phenotype of human peripheral blood T cells and TIL. Tumor-specific T cells with disrupted Cish function were adoptively transferred into tumor-bearing mice and evaluated for efficacy with or without checkpoint blockade. FINDINGS: CISH expression was associated with T cell dysfunction. CISH deletion using CRISPR/Cas9 resulted in hyper-activation and improved functional avidity against tumor-derived neoantigens without perturbing T cell maturation. Cish knockout resulted in increased susceptibility to checkpoint blockade in vivo. CONCLUSIONS: CISH negatively regulates human T cell effector function, and its genetic disruption offers a novel avenue to improve the therapeutic efficacy of adoptive TIL therapy. FUNDING: This study was funded by Intima Bioscience, U.S. and in part through the Intramural program CCR at the National Cancer Institute.


Lymphocytes, Tumor-Infiltrating , T-Lymphocytes , Adoptive Transfer , Animals , Cytokines/metabolism , Humans , Immunotherapy, Adoptive/methods , Mice
4.
Nat Commun ; 10(1): 5222, 2019 11 19.
Article En | MEDLINE | ID: mdl-31745080

The fusion of genome engineering and adoptive cellular therapy holds immense promise for the treatment of genetic disease and cancer. Multiplex genome engineering using targeted nucleases can be used to increase the efficacy and broaden the application of such therapies but carries safety risks associated with unintended genomic alterations and genotoxicity. Here, we apply base editor technology for multiplex gene modification in primary human T cells in support of an allogeneic CAR-T platform and demonstrate that base editor can mediate highly efficient multiplex gene disruption with minimal double-strand break induction. Importantly, multiplex base edited T cells exhibit improved expansion and lack double strand break-induced translocations observed in T cells edited with Cas9 nuclease. Our findings highlight base editor as a powerful platform for genetic modification of therapeutically relevant primary cell types.


CRISPR-Cas Systems , Cell Engineering/methods , DNA Breaks, Double-Stranded , Gene Editing/methods , T-Lymphocytes/metabolism , Cells, Cultured , High-Throughput Nucleotide Sequencing/methods , Humans , Immunotherapy, Adoptive/methods , Reproducibility of Results , T-Lymphocytes/cytology
5.
Haematologica ; 101(10): 1190-1199, 2016 10.
Article En | MEDLINE | ID: mdl-27418650

Juvenile myelomonocytic leukemia is a rare myeloproliferative neoplasm characterized by hyperactive RAS signaling. Neurofibromin1 (encoded by the NF1 gene) is a negative regulator of RAS activation. Patients with neurofibromatosis type 1 harbor loss-of-function mutations in NF1 and have a 200- to 500-fold increased risk of juvenile myelomonocytic leukemia. Leukemia cells from patients with juvenile myelomonocytic leukemia display hypersensitivity to certain cytokines, such as granulocyte-macrophage colony-stimulating factor. The granulocyte-macrophage colony-stimulating factor receptor utilizes pre-associated JAK2 to initiate signals after ligand binding. JAK2 subsequently activates STAT5, among other downstream effectors. Although STAT5 is gaining recognition as an important mediator of growth factor signaling in myeloid leukemias, the contribution of STAT5 to the development of hyperactive RAS-initiated myeloproliferative disease has not been well described. In this study, we investigated the consequence of STAT5 attenuation via genetic and pharmacological approaches in Nf1-deficient murine models of juvenile myelomonocytic leukemia. We found that homozygous Stat5 deficiency extended the lifespan of Nf1-deficient mice and eliminated the development of myeloproliferative neoplasm associated with Nf1 gene loss. Likewise, we found that JAK inhibition with ruxolitinib attenuated myeloproliferative neoplasm in Nf1-deficient mice. Finally, we found that primary cells from a patient with KRAS-mutant juvenile myelomonocytic leukemia displayed reduced colony formation in response to JAK2 inhibition. Our findings establish a central role for STAT5 activation in the pathogenesis of juvenile myelomonocytic leukemia and suggest that targeting this pathway may be of clinical utility in these patients.


Janus Kinase 2/metabolism , Leukemia, Myelomonocytic, Juvenile/etiology , Myeloproliferative Disorders/etiology , Neurofibromin 1/deficiency , STAT5 Transcription Factor/physiology , Animals , Disease Models, Animal , Humans , Leukemia, Myeloid/etiology , Leukemia, Myeloid/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Mice , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction
6.
Blood ; 124(22): 3274-83, 2014 Nov 20.
Article En | MEDLINE | ID: mdl-25316678

Mutant RAS oncoproteins activate signaling molecules that drive oncogenesis in multiple human tumors including acute myelogenous leukemia (AML). However, the specific functions of these pathways in AML are unclear, thwarting the rational application of targeted therapeutics. To elucidate the downstream functions of activated NRAS in AML, we used a murine model that harbors Mll-AF9 and a tetracycline-repressible, activated NRAS (NRAS(G12V)). Using computational approaches to explore our gene-expression data sets, we found that NRAS(G12V) enforced the leukemia self-renewal gene-expression signature and was required to maintain an MLL-AF9- and Myb-dependent leukemia self-renewal gene-expression program. NRAS(G12V) was required for leukemia self-renewal independent of its effects on growth and survival. Analysis of the gene-expression patterns of leukemic subpopulations revealed that the NRAS(G12V)-mediated leukemia self-renewal signature is preferentially expressed in the leukemia stem cell-enriched subpopulation. In a multiplexed analysis of RAS-dependent signaling, Mac-1(Low) cells, which harbor leukemia stem cells, were preferentially sensitive to NRAS(G12V) withdrawal. NRAS(G12V) maintained leukemia self-renewal through mTOR and MEK pathway activation, implicating these pathways as potential targets for cancer stem cell-specific therapies. Together, these experimental results define a RAS oncogene-driven function that is critical for leukemia maintenance and represents a novel mechanism of oncogene addiction.


Cell Proliferation/genetics , GTP Phosphohydrolases/physiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/physiology , Amino Acid Substitution , Animals , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Leukemic , Glycine/genetics , Humans , Membrane Proteins/genetics , Mice , Mice, SCID , Oncogenes/physiology , Transcriptome , Tumor Cells, Cultured , Valine/genetics
7.
Blood ; 119(19): 4512-23, 2012 May 10.
Article En | MEDLINE | ID: mdl-22427200

Patients with a t(9;11) translocation (MLL-AF9) develop acute myeloid leukemia (AML), and while in mice the expression of this fusion oncogene also results in the development of myeloid leukemia, it is with long latency. To identify mutations that cooperate with Mll-AF9, we infected neonatal wild-type (WT) or Mll-AF9 mice with a murine leukemia virus (MuLV). MuLV-infected Mll-AF9 mice succumbed to disease significantly faster than controls presenting predominantly with myeloid leukemia while infected WT animals developed predominantly lymphoid leukemia. We identified 88 candidate cancer genes near common sites of proviral insertion. Analysis of transcript levels revealed significantly elevated expression of Mn1, and a trend toward increased expression of Bcl11a and Fosb in Mll-AF9 murine leukemia samples with proviral insertions proximal to these genes. Accordingly, FOSB and BCL11A were also overexpressed in human AML harboring MLL gene translocations. FOSB was revealed to be essential for growth in mouse and human myeloid leukemia cells using shRNA lentiviral vectors in vitro. Importantly, MN1 cooperated with Mll-AF9 in leukemogenesis in an in vivo BM viral transduction and transplantation assay. Together, our data identified genes that define transcription factor networks and important genetic pathways acting during progression of leukemia induced by MLL fusion oncogenes.


Cell Transformation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Leukemia/genetics , Mutagenesis, Insertional , Myeloid-Lymphoid Leukemia Protein/physiology , Oncogene Proteins, Fusion/physiology , Animals , Animals, Newborn , Cells, Cultured , DNA Mutational Analysis/methods , Disease Models, Animal , HEK293 Cells , Humans , Leukemia/pathology , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional/physiology , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , U937 Cells
8.
Am J Hematol ; 86(7): 579-85, 2011 Jul.
Article En | MEDLINE | ID: mdl-21681782

Juvenile Myelomonocytic Leukemia (JMML) is a relentlessly progressive myeloproliferative/myelodysplastic (MPD/MDS) hematopoietic disorder more common in patients with any one of at least three distinct genetic lesions, specifically NF1 gene loss and PTPN11 and NRAS mutations. NF1 and PTPN11 are molecular lesions associated with Neurofibromatosis Syndrome Type I (NF1 Syndrome) and Noonan's Syndrome, respectively. The occurrence of JMML is rare; even among those predisposed with these syndromes to development of disease, and secondary genetic events likely contribute to the development and progression of disease. In NF1 syndrome, loss of p53 function is a common event in solid tumors, but uncommon in JMML, suggesting that the p53 pathway may be modified by other events in this hematopoietic disorder. The work presented here investigates the possible role of the p19(Arf) (p19) tumor suppressor in development of MPD associated with Nf1 gene loss in mice. We find that Nf1 mutant hematopoietic cells with loss of p19 develop accelerated hematopoietic disease similar to acute leukemia with a variable phenotype. This suggests that p19 may play a role in development of JMML and evaluation of the human p19 homolog (p14(ARF)) in JMML may be informative.


Cyclin-Dependent Kinase Inhibitor p16/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Neurofibromin 1/genetics , Acute Disease , Animals , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Leukemia, Myelomonocytic, Juvenile/metabolism , Leukemia, Myelomonocytic, Juvenile/pathology , Mice , Mice, Mutant Strains , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/pathology , Neurofibromin 1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Cancer Res ; 69(21): 8429-37, 2009 Nov 01.
Article En | MEDLINE | ID: mdl-19843846

The Sleeping Beauty (SB) transposon system has been used as a somatic mutagen to identify candidate cancer genes. In previous studies, efficient leukemia/lymphoma formation on an otherwise wild-type genetic background occurred in mice undergoing whole-body mobilization of transposons, but was accompanied by high levels of embryonic lethality. To explore the utility of SB for large-scale cancer gene discovery projects, we have generated mice that carry combinations of different transposon and transposase transgenes. We have identified a transposon/transposase combination that promotes highly penetrant leukemia/lymphoma formation on an otherwise wild-type genetic background, yet does not cause embryonic lethality. Infiltrating gliomas also occurred at lower penetrance in these mice. SB-induced or accelerated tumors do not harbor large numbers of chromosomal amplifications or deletions, indicating that transposon mobilization likely promotes tumor formation by insertional mutagenesis of cancer genes, and not by promoting wide-scale genomic instability. Cloning of transposon insertions from lymphomas/leukemias identified common insertion sites at known and candidate novel cancer genes. These data indicate that a high mutagenesis rate can be achieved using SB without high levels of embryonic lethality or genomic instability. Furthermore, the SB system could be used to identify new genes involved in lymphomagenesis/leukemogenesis.


DNA Transposable Elements/genetics , Embryo, Mammalian/cytology , Genes, Lethal , Glioma/genetics , Leukemia/genetics , Lymphoma/genetics , Transposases/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Comparative Genomic Hybridization , Embryo, Mammalian/metabolism , Flow Cytometry , Gene Expression Profiling , Genomic Instability , Glioma/pathology , Immunoenzyme Techniques , Leukemia/pathology , Lymphoma/pathology , Mice , Mice, Transgenic , Mutagenesis , Oligonucleotide Array Sequence Analysis , Survival Rate
10.
Blood ; 113(5): 1086-96, 2009 Jan 29.
Article En | MEDLINE | ID: mdl-18952898

To study the oncogenic role of the NRAS oncogene (NRAS(G12V)) in the context of acute myeloid leukemia (AML), we used a Vav promoter-tetracycline transactivator (Vav-tTA)-driven repressible TRE-NRAS(G12V) transgene system in Mll-AF9 knock-in mice developing AML. Conditional repression of NRAS(G12V) expression greatly reduced peripheral white blood cell (WBC) counts in leukemia recipient mice and induced apoptosis in the transplanted AML cells correlated with reduced Ras/Erk signaling. After marked decrease of AML blast cells, myeloproliferative disease (MPD)-like AML relapsed characterized by cells that did not express NRAS(G12V). In comparison with primary AML, the MPD-like AML showed significantly reduced aggressiveness, reduced myelosuppression, and a more differentiated phenotype. We conclude that, in AML induced by an Mll-AF9 transgene, NRAS(G12V) expression contributes to acute leukemia maintenance by suppressing apoptosis and reducing differentiation of leukemia cells. Moreover, NRAS(G12V) oncogene has a cell nonautonomous role in suppressing erythropoiesis that results in the MPD-like AML show significantly reduced ability to induce anemia. Our results imply that targeting NRAS or RAS oncogene-activated pathways is a good therapeutic strategy for AML and attenuating aggressiveness of relapsed AML.


Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Amino Acid Substitution , Animals , Apoptosis/genetics , Erythropoiesis/genetics , Gene Knock-In Techniques , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Leukocyte Count , Mice , Mice, Transgenic , Mutation, Missense , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Transplantation , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Recurrence
11.
Blood ; 109(4): 1687-91, 2007 Feb 15.
Article En | MEDLINE | ID: mdl-17090653

Neurofibromatosis type 1 (NF1) syndrome is caused by germline mutations in the NF1 tumor suppressor, which encodes neurofibromin, a GTPase activating protein for Ras. Children with NF1 are predisposed to juvenile myelomonocytic leukemia (JMML) and lethally irradiated mice given transplants with homozygous Nf1 mutant (Nf1-/-) hematopoietic stem cells develop a fatal myeloproliferative disorder (MPD) that models JMML. We investigated the requirement for signaling through the GM-CSF receptor to initiate and sustain this MPD by generating Nf1 mutant hematopoietic cells lacking the common beta chain (Beta c) of the GM-CSF receptor. Mice reconstituted with Nf1-/-, beta c-/- stem cells did not develop evidence of MPD despite the presence of increased number of immature hematopoietic progenitors in the bone marrow. Interestingly, when the Mx1-Cre transgene was used to inactivate a conditional Nf1 mutant allele in hematopoietic cells, concomitant loss of beta c-/- reduced the severity of the MPD, but did not abrogate it. Whereas inhibiting GM-CSF signaling may be of therapeutic benefit in JMML, our data also demonstrate aberrant proliferation of Nf1-/-myeloid progenitors that is independent of signaling through the GM-CSF receptor.


Leukemia, Myelomonocytic, Chronic/etiology , Myeloproliferative Disorders/etiology , Neurofibromatosis 1/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Disease Models, Animal , Hematopoietic Stem Cell Transplantation , Leukemia, Myelomonocytic, Chronic/prevention & control , Mice , Mice, Mutant Strains , Myeloproliferative Disorders/prevention & control , Signal Transduction
12.
J Pharmacol Exp Ther ; 314(2): 668-74, 2005 Aug.
Article En | MEDLINE | ID: mdl-15857949

Methotrexate (MTX) is an effective antitumor agent that has been demonstrated to be particularly useful in the treatment of hematopoietic neoplasms but causes substantial hematologic and gastrointestinal toxicity. We previously demonstrated that transplantation with transgenic marrow expressing drug-resistant dihydrofolate reductase (DHFR) into animals preconditioned by irradiation substantially protected recipient mice from the toxic side effects of methotrexate administration. Here we test the use of methotrexate itself as a preconditioning agent for engraftment of drug-resistant transgenic marrow, subsequently conferring drug resistance upon recipient animals. Administration of methotrexate beginning 1 or 2 weeks prior to or on the same day as transplantation with drug-resistant DHFR transgenic marrow did not allow sufficient engraftment to confer drug resistance to most unirradiated recipients. A small number of animals were curiously protected from lethal MTX toxicity but exhibited extremely low hematocrits and were not engrafted with stem cells, as indicated by low engraftment levels assessed in secondary transplant recipients. However, we subsequently found that MTX preconditioning allowed sufficient engraftment of DHFR transgenic marrow to confer drug resistance if MTX administration was withdrawn at the time of bone marrow transplantation (BMT) and withheld until 2 weeks post-transplant. Quantitative molecular analysis of primary and secondary recipients indicated a stem cell engraftment level of approximately 1%, consistent with previous studies demonstrating that a low level of DHFR transgenic cell engraftment was sufficient to confer drug resistance in recipient animals. We conclude that MTX can be used as a preconditioning agent for subsequent engraftment of hematopoietic stem cells, in this case conferring resistance to MTX.


Bone Marrow Transplantation/physiology , Folic Acid Antagonists/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Immunosuppressive Agents/pharmacology , Methotrexate/pharmacology , Tetrahydrofolate Dehydrogenase/metabolism , Animals , Blotting, Southern , Bone Marrow Transplantation/immunology , Drug Resistance , Hematocrit , Hematopoiesis/drug effects , In Situ Hybridization , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction , Tetrahydrofolate Dehydrogenase/genetics
13.
J Pharmacol Exp Ther ; 300(3): 1075-84, 2002 Mar.
Article En | MEDLINE | ID: mdl-11861818

Expression of drug-resistant forms of dihydrofolate reductase (DHFR) in hematopoietic cells confers substantial resistance of animals to antifolate administration. In this study, we tested whether the chemoprotection conferred by expression of the tyrosine-22 variant DHFR could be used for more effective therapy of the 32Dp210 murine model of chronic myeloid leukemia (CML). 32Dp210 tumor cells were found to be sensitive to methotrexate (MTX) in vitro, whereas cells expressing the tyrosine-22 DHFR gene were protected from MTX at up to micromolar concentrations. MTX administered at low dose (2 mg/kg/day) did not protect normal C3H-He/J mice from 32Dp210 tumor infused intravenously, with drug toxicity limiting the administration of higher doses. Animals engrafted with transgenic tyrosine-22 DHFR marrow were protected from greater MTX doses (up to 6 mg/kg/day). However, the increased doses of MTX afforded by drug-resistance gene expression surprisingly resulted in decreased survival of the transplanted tumor-bearing animals, with increased levels of tumor detected in peripheral blood. This apparent exacerbation of tumor progression by MTX was not observed in DHFR transgenic mice in which all cells and tissues contain the drug-resistance gene. This suggests that increased tumor progression in MTX-administered animals resulted from MTX sensitivity of a nonhematopoietic host component, thus allowing tumor expansion. We conclude that MTX exacerbates tumor progression in the 32Dp210 model of CML, and that based on this model alternate DHFR inhibitors combined with drug-resistant DHFR or other chemotherapeutic agent/drug-resistance gene combinations may be required for the application of drug-resistance gene expression to the treatment of CML.


Folic Acid Antagonists/toxicity , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Methotrexate/toxicity , Animals , Bone Marrow Transplantation , Cell Survival , Disease Progression , Drug Resistance, Neoplasm , Flow Cytometry , Hematocrit , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mice , Mice, Inbred C3H , Organ Size/drug effects , Survival Analysis , Tetrahydrofolate Dehydrogenase/metabolism , Tumor Cells, Cultured
...