8 results on '"Ryan, Theresa"'
Search Results
2. Cytosine base editing enables quadruple-edited allogeneic CART cells for T-ALL.
- Author
-
Diorio C, Murray R, Naniong M, Barrera L, Camblin A, Chukinas J, Coholan L, Edwards A, Fuller T, Gonzales C, Grupp SA, Ladd A, Le M, Messana A, Musenge F, Newman H, Poh YC, Poulin H, Ryan T, Shraim R, Tasian SK, Vincent T, Young L, Zhang Y, Ciaramella G, Gehrke J, and Teachey DT
- Subjects
- CRISPR-Cas Systems, Cytosine, Gene Editing methods, Humans, Hematopoietic Stem Cell Transplantation, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Allogeneic chimeric antigen receptor T-cell (CART) therapies require multiple gene edits to be clinically tractable. Most allogeneic CARTs have been created using gene editing techniques that induce DNA double-stranded breaks (DSBs), resulting in unintended on-target editing outcomes with potentially unforeseen consequences. Cytosine base editors (CBEs) install C•G to T•A point mutations in T cells, with between 90% and 99% efficiency to silence gene expression without creating DSBs, greatly reducing or eliminating undesired editing outcomes following multiplexed editing as compared with clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9). Using CBE, we developed 7CAR8, a CD7-directed allogeneic CART created using 4 simultaneous base edits. We show that CBE, unlike CRISPR-Cas9, does not impact T-cell proliferation, lead to aberrant DNA damage response pathway activation, or result in karyotypic abnormalities following multiplexed editing. We demonstrate 7CAR8 to be highly efficacious against T-cell acute lymphoblastic leukemia (T-ALL) using multiple in vitro and in vivo models. Thus, CBE is a promising technology for applications requiring multiplexed gene editing and can be used to manufacture quadruple-edited 7CAR8 cells, with high potential for clinical translation for relapsed and refractory T-ALL., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
3. Potent efficacy of combined PI3K/mTOR and JAK or ABL inhibition in murine xenograft models of Ph-like acute lymphoblastic leukemia.
- Author
-
Tasian SK, Teachey DT, Li Y, Shen F, Harvey RC, Chen IM, Ryan T, Vincent TL, Willman CL, Perl AE, Hunger SP, Loh ML, Carroll M, and Grupp SA
- Subjects
- Animals, Cell Proliferation drug effects, Humans, Janus Kinases antagonists & inhibitors, Mice, Phosphoinositide-3 Kinase Inhibitors, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Random Allocation, TOR Serine-Threonine Kinases antagonists & inhibitors, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Protein Kinase Inhibitors pharmacology, Signal Transduction drug effects
- Abstract
Philadelphia chromosome (Ph)-like B-cell acute lymphoblastic leukemia (Ph-like ALL) is associated with activated JAK/STAT, Abelson kinase (ABL), and/or phosphatidylinositol 3-kinase (PI3K) signaling and poor clinical outcomes. PI3K pathway signaling inhibitors have been minimally investigated in Ph-like ALL. We hypothesized that targeted inhibition of PI3Kα, PI3Kδ, PI3K/mTOR, or target of rapamycin complex 1/2 (TORC1/TORC2) would decrease leukemia proliferation and abrogate aberrant kinase signaling and that combined PI3K pathway and JAK inhibition or PI3K pathway and SRC/ABL inhibition would have superior efficacy compared to inhibitor monotherapy. We treated 10 childhood ALL patient-derived xenograft models harboring various Ph-like genomic alterations with 4 discrete PI3K pathway protein inhibitors and observed marked leukemia reduction and in vivo signaling inhibition in all models. Treatment with dual PI3K/mTOR inhibitor gedatolisib resulted in near eradication of ALL in cytokine receptor-like factor 2 (CRLF2)/JAK-mutant models with mean 92.2% (range, 86.0%-99.4%) reduction vs vehicle controls (P < .0001) and in prolonged animal survival. Gedatolisib also inhibited ALL proliferation in ABL/platelet-derived growth factor receptor (PDGFR)-mutant models with mean 66.9% (range, 42.0%-87.6%) reduction vs vehicle (P < .0001). Combined gedatolisib and ruxolitinib treatment of CRLF2/JAK-mutant models more effectively inhibited ALL proliferation than either inhibitor alone (P < .001) and further enhanced survival. Similarly, superior efficacy of combined gedatolisib and dasatinib was observed in ABL/PDGFR-mutant models (P < .001). Overall, PI3K/mTOR inhibition potently decreased ALL burden in vivo; antileukemia activity was further enhanced with combination inhibitor therapy. Clinical trials testing combinations of kinase inhibitors in Ph-like ALL patients are indicated., (© 2017 by The American Society of Hematology.)
- Published
- 2017
- Full Text
- View/download PDF
4. Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic leukemia.
- Author
-
Maude SL, Dolai S, Delgado-Martin C, Vincent T, Robbins A, Selvanathan A, Ryan T, Hall J, Wood AC, Tasian SK, Hunger SP, Loh ML, Mullighan CG, Wood BL, Hermiston ML, Grupp SA, Lock RB, and Teachey DT
- Subjects
- Adolescent, Animals, Child, Child, Preschool, Female, Humans, Interleukin-7 metabolism, Janus Kinases genetics, Male, Mice, Mice, Inbred NOD, Mice, SCID, Mutation, Nitriles, Precursor Cells, T-Lymphoid drug effects, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-bcl-2 metabolism, Pyrazoles pharmacology, Pyrimidines, STAT Transcription Factors genetics, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Young Adult, Janus Kinases antagonists & inhibitors, Precursor Cells, T-Lymphoid metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, STAT Transcription Factors antagonists & inhibitors
- Abstract
Early T-cell precursor (ETP) acute lymphoblastic leukemia (ALL) is a recently described subtype of T-ALL characterized by a unique immunophenotype and genomic profile, as well as a high rate of induction failure. Frequent mutations in cytokine receptor and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathways led us to hypothesize that ETP-ALL is dependent on JAK/STAT signaling. Here we demonstrate aberrant activation of the JAK/STAT pathway in ETP-ALL blasts relative to non-ETP T-ALL. Moreover, ETP-ALL showed hyperactivation of STAT5 in response to interleukin-7, an effect that was abrogated by the JAK1/2 inhibitor ruxolitinib. In vivo, ruxolitinib displayed activity in 6 of 6 patient-derived murine xenograft models of ETP-ALL, with profound single-agent efficacy in 5 models. Ruxolitinib treatment decreased peripheral blast counts relative to pretreatment levels and compared with control (P < .01) in 5 of 6 ETP-ALL xenografts, with marked reduction in mean splenic blast counts (P < .01) in 6 of 6 samples. Surprisingly, both JAK/STAT pathway activation and ruxolitinib efficacy were independent of the presence of JAK/STAT pathway mutations, raising the possibility that the therapeutic potential of ruxolitinib in ETP-ALL extends beyond those cases with JAK mutations. These findings establish the preclinical in vivo efficacy of ruxolitinib in ETP-ALL, a biologically distinct subtype for which novel therapies are needed., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
5. mTOR inhibitors are synergistic with methotrexate: an effective combination to treat acute lymphoblastic leukemia.
- Author
-
Teachey DT, Sheen C, Hall J, Ryan T, Brown VI, Fish J, Reid GS, Seif AE, Norris R, Chang YJ, Carroll M, and Grupp SA
- Subjects
- Animals, Cell Line, Tumor, Cyclin D, Cyclins metabolism, Drug Resistance, Neoplasm, Drug Synergism, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Transplantation, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, TOR Serine-Threonine Kinases, Tetrahydrofolate Dehydrogenase metabolism, Transplantation, Heterologous, Antimetabolites, Antineoplastic administration & dosage, Methotrexate administration & dosage, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Protein Kinase Inhibitors administration & dosage, Protein Kinases metabolism
- Abstract
We have previously demonstrated that mTOR inhibitors (MTIs) are active in preclinical models of acute lymphoblastic leukemia (ALL). MTIs may increase degradation of cyclin D1, a protein involved in dihydrofolate reductase (DHFR) synthesis. Because resistance to methotrexate may correlate with high DHFR expression, we hypothesized MTIs may increase sensitivity of ALL to methotrexate through decreasing DHFR by increasing turn-over of cyclin D1. We tested this hypothesis using multiple ALL cell lines and nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice xenografted with human ALL. We found MTIs and methotrexate were synergistic in combination in vitro and in vivo. Mice treated with both drugs went into a complete and durable remission whereas single agent treatment caused an initial partial response that ultimately progressed. ALL cells treated with MTIs had markedly decreased expression of DHFR and cyclin D1, providing a novel mechanistic explanation for a combined effect. We found methotrexate and MTIs are an effective and potentially synergistic combination in ALL.
- Published
- 2008
- Full Text
- View/download PDF
6. Targeting Notch signaling in autoimmune and lymphoproliferative disease.
- Author
-
Teachey DT, Seif AE, Brown VI, Bruno M, Bunte RM, Chang YJ, Choi JK, Fish JD, Hall J, Reid GS, Ryan T, Sheen C, Zweidler-McKay P, and Grupp SA
- Subjects
- Animals, Antibodies, Antinuclear blood, Antibodies, Antinuclear immunology, Dipeptides adverse effects, Dipeptides therapeutic use, Disease Models, Animal, Drug Evaluation, Preclinical, Enzyme Inhibitors adverse effects, Enzyme Inhibitors therapeutic use, Humans, Immunoglobulin G blood, Immunoglobulin G immunology, Lupus Erythematosus, Systemic blood, Lupus Erythematosus, Systemic diagnostic imaging, Lupus Erythematosus, Systemic immunology, Lymph Nodes diagnostic imaging, Lymph Nodes immunology, Lymph Nodes metabolism, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Lymphoproliferative Disorders blood, Lymphoproliferative Disorders diagnostic imaging, Lymphoproliferative Disorders immunology, Mice, Mice, Inbred CBA, Mice, Inbred MRL lpr, Nephritis blood, Nephritis diagnostic imaging, Nephritis drug therapy, Nephritis immunology, Random Allocation, Receptors, Notch immunology, Signal Transduction immunology, Spleen diagnostic imaging, Spleen immunology, Spleen metabolism, T-Lymphocytes metabolism, Ultrasonography, Dipeptides pharmacology, Enzyme Inhibitors pharmacology, Lupus Erythematosus, Systemic drug therapy, Lymphoproliferative Disorders drug therapy, Receptors, Notch antagonists & inhibitors, Signal Transduction drug effects, T-Lymphocytes immunology
- Abstract
Patients with autoimmune lymphoproliferative syndrome (ALPS) and systemic lupus erythematosis (SLE) have T-cell dysregulation and produce abnormal, activated T lymphocytes and an atypical peripheral T-cell population, termed double negative T cells (DNTs). T-cell functions, including DNT transition in T-cell development and T-cell activation, are critically dependent on Notch signaling. We hypothesized that inhibiting Notch signaling would be effective in ALPS and SLE by reducing the production of abnormal DNTs and by blocking aberrant T-cell activation. We tested this hypothesis using murine models of ALPS and SLE. Mice were randomized to treatment with the notch pathway inhibitor (gamma-secretase inhibitor), N-S-phenyl-glycine-t-butyl ester (DAPT), or vehicle control. Response to treatment was assessed by measurement of DNTs in blood and lymphoid tissue, by monitoring lymph node and spleen size with ultrasound, by quantifying cytokines by bead-array, by ELISA for total IgG and anti-double-stranded DNA (dsDNA) specific antibodies, and by histopathologic assessment for nephritis. We found a profound and statistically significant decrease in all disease parameters, comparing DAPT-treated mice to controls. Using a novel dosing schema, we avoided the reported toxicities of gamma-secretase inhibitors. Inhibiting the Notch signaling pathway may thus present an effective, novel, and well-tolerated treatment for autoimmune and lymphoproliferative diseases.
- Published
- 2008
- Full Text
- View/download PDF
7. Hypercortisolism and Brain Ventricular Size in Normal Aging and Alzheimer's Disease.
- Author
-
Lawlor BA, Mohs RC, Losonczy M, Ryan TM, Guo L, Davis KL, Bierer LM, and Schmeidler J
- Published
- 1994
- Full Text
- View/download PDF
8. Response to Combination Cholinergic/Noradrenergic Treatments in a Patient With Alzheimer's Disease.
- Author
-
Aisen PS, Bierer LM, Davidson M, Ryan TM, Kaminsky R, and Davis KL
- Abstract
A patient with probable Alzheimer's disease participated in four studies involving cholinergic and combination cholinergic/noradrenergic treatment regimens. This patient demonstrated clinically significant improvement only with the combination of physostigmine and clonidine. The implications of these findings are discussed with regard to strategies for neurotransmitter augmentation therapy for Alzheimer's disease., (Copyright © 1993 American Association for Geriatric Psychiatry. Published by Elsevier Inc. All rights reserved.)
- Published
- 1993
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.