85 results on '"Eide CA"'
Search Results
2. Persistent LYN signaling in imatinib-resistant, BCR-ABL-independent chronic myelogenous leukemia.
- Author
-
O'Hare T, Eide CA, and Deiniger MW
- Published
- 2008
- Full Text
- View/download PDF
3. New strategies for the first-line treatment of chronic myeloid leukemia: can resistance be avoided?
- Author
-
Snead JL, O'Hare T, Eide CA, and Deininger MW
- Published
- 2008
4. Overcoming clinical BCR-ABL1 compound mutant resistance with combined ponatinib and asciminib therapy.
- Author
-
Eide CA, Brewer D, Xie T, Schultz AR, Savage SL, Muratcioglu S, Merz N, Press RD, O'Hare T, Jacob T, Vu TQ, Tognon CE, Macey TA, Kuriyan J, Kalodimos CG, and Druker BJ
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Male, Female, Niacinamide analogs & derivatives, Pyrazoles, Pyridazines therapeutic use, Pyridazines pharmacology, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl antagonists & inhibitors, Imidazoles pharmacology, Imidazoles therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Drug Resistance, Neoplasm genetics, Drug Resistance, Neoplasm drug effects, Mutation, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology
- Abstract
BCR-ABL1 compound mutations can lead to resistance to ABL1 inhibitors in chronic myeloid leukemia (CML), which could be targeted by combining the ATP-site inhibitor ponatinib and the allosteric inhibitor asciminib. Here, we report the clinical validation of this approach in a CML patient, providing a basis for combination therapy to overcome such resistance., Competing Interests: Declaration of interests B.J.D. serves on scientific advisory boards for Aileron Therapeutics, Therapy Architects (ALLCRON), Cepheid, Vivid Biosciences, Celgene, RUNX1 Research Program, Novartis, Gilead Sciences (inactive), and Monojul (inactive); serves on scientific advisory boards and receives stock from Aptose Biosciences, Blueprint Medicines, EnLiven Therapeutics, Iterion Therapeutics, Third Coast Therapeutics, and GRAIL (inactive on scientific advisory board); is scientific founder of MolecularMD (inactive, acquired by ICON); serves on the board of directors and receives stock from Amgen and Vincera Pharma; serves on the board of directors for Burroughs Wellcome Fund and CureOne; serves on the joint steering committee for Beat AML LLS; is founder of VB Therapeutics; has a sponsored research agreement with EnLiven Therapeutics; receives clinical trial funding from Novartis, Bristol-Myers Squibb, and Pfizer., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Targeting CCL2/CCR2 Signaling Overcomes MEK Inhibitor Resistance in Acute Myeloid Leukemia.
- Author
-
Modak RV, de Oliveira Rebola KG, McClatchy J, Mohammadhosseini M, Damnernsawad A, Kurtz SE, Eide CA, Wu G, Laderas T, Nechiporuk T, Gritsenko MA, Hansen JR, Hutchinson C, Gosline SJC, Piehowski P, Bottomly D, Short N, Rodland K, McWeeney SK, Tyner JW, and Agarwal A
- Subjects
- Humans, Cell Line, Tumor, Cell Proliferation drug effects, Animals, Pyridones pharmacology, Pyridones therapeutic use, Mice, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Receptors, CCR2 metabolism, Receptors, CCR2 antagonists & inhibitors, Receptors, CCR2 genetics, Drug Resistance, Neoplasm genetics, Chemokine CCL2 metabolism, Chemokine CCL2 genetics, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Signal Transduction drug effects
- Abstract
Purpose: Emerging evidence underscores the critical role of extrinsic factors within the microenvironment in protecting leukemia cells from therapeutic interventions, driving disease progression, and promoting drug resistance in acute myeloid leukemia (AML). This finding emphasizes the need for the identification of targeted therapies that inhibit intrinsic and extrinsic signaling to overcome drug resistance in AML., Experimental Design: We performed a comprehensive analysis utilizing a cohort of ∼300 AML patient samples. This analysis encompassed the evaluation of secreted cytokines/growth factors, gene expression, and ex vivo drug sensitivity to small molecules. Our investigation pinpointed a notable association between elevated levels of CCL2 and diminished sensitivity to the MEK inhibitors (MEKi). We validated this association through loss-of-function and pharmacologic inhibition studies. Further, we deployed global phosphoproteomics and CRISPR/Cas9 screening to identify the mechanism of CCR2-mediated MEKi resistance in AML., Results: Our multifaceted analysis unveiled that CCL2 activates multiple prosurvival pathways, including MAPK and cell-cycle regulation in MEKi-resistant cells. Employing combination strategies to simultaneously target these pathways heightened growth inhibition in AML cells. Both genetic and pharmacologic inhibition of CCR2 sensitized AML cells to trametinib, suppressing proliferation while enhancing apoptosis. These findings underscore a new role for CCL2 in MEKi resistance, offering combination therapies as an avenue to circumvent this resistance., Conclusions: Our study demonstrates a compelling rationale for translating CCL2/CCR2 axis inhibitors in combination with MEK pathway-targeting therapies, as a potent strategy for combating drug resistance in AML. This approach has the potential to enhance the efficacy of treatments to improve AML patient outcomes., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
6. Clinical Correlates of Venetoclax-Based Combination Sensitivities to Augment Acute Myeloid Leukemia Therapy.
- Author
-
Eide CA, Kurtz SE, Kaempf A, Long N, Joshi SK, Nechiporuk T, Huang A, Dibb CA, Taylor A, Bottomly D, McWeeney SK, Minnier J, Lachowiez CA, Saultz JN, Swords RT, Agarwal A, Chang BH, Druker BJ, and Tyner JW
- Subjects
- Humans, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Azacitidine pharmacology, Azacitidine therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use
- Abstract
The BCL2 inhibitor venetoclax combined with the hypomethylating agent azacytidine shows significant clinical benefit in a subset of patients with acute myeloid leukemia (AML); however, resistance limits response and durability. We prospectively profiled the ex vivo activity of 25 venetoclax-inclusive combinations on primary AML patient samples to identify those with improved potency and synergy compared with venetoclax + azacytidine (Ven + azacytidine). Combination sensitivities correlated with tumor cell state to discern three patterns: primitive selectivity resembling Ven + azacytidine, monocytic selectivity, and broad efficacy independent of cell state. Incorporation of immunophenotype, mutation, and cytogenetic features further stratified combination sensitivity for distinct patient subtypes. We dissect the biology underlying the broad, cell state-independent efficacy for the combination of venetoclax plus the JAK1/2 inhibitor ruxolitinib. Together, these findings support opportunities for expanding the impact of venetoclax-based drug combinations in AML by leveraging clinical and molecular biomarkers associated with ex vivo responses., Significance: By mapping drug sensitivity data to clinical features and tumor cell state, we identify novel venetoclax combinations targeting patient subtypes who lack sensitivity to Ven + azacytidine. This provides a framework for a taxonomy of AML informed by readily available sets of clinical and genetic features obtained as part of standard care. See related commentary by Becker, p. 437 . This article is featured in Selected Articles from This Issue, p. 419., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
7. Patient-Specific Targeting of the T-Cell Receptor Variable Region as a Therapeutic Strategy in Clonal T-Cell Diseases.
- Author
-
Lucero OM, Lee JA, Bowman J, Johnson K, Sapparapu G, Thomas JK, Fan G, Chang BH, Thiel-Klare K, Eide CA, Okada C, Palazzolo M, Lind E, Kosaka Y, Druker BJ, Lydon N, and Bowers PM
- Subjects
- Humans, Mice, Animals, Rituximab, T-Lymphocytes immunology, Receptors, Antigen, T-Cell, alpha-beta genetics, Receptors, Antigen, T-Cell, alpha-beta immunology, Receptors, Antigen, T-Cell genetics, Lymphoma, T-Cell
- Abstract
Purpose: Targeted therapeutics are a goal of medicine. Methods for targeting T-cell lymphoma lack specificity for the malignant cell, leading to elimination of healthy cells. The T-cell receptor (TCR) is designed for antigen recognition. T-cell malignancies expand from a single clone that expresses one of 48 TCR variable beta (Vβ) genes, providing a distinct therapeutic target. We hypothesized that a mAb that is exclusive to a specific Vβ would eliminate the malignant clone while having minimal effects on healthy T cells., Experimental Design: We identified a patient with large granular T-cell leukemia and sequenced his circulating T-cell population, 95% of which expressed Vβ13.3. We developed a panel of anti-Vβ13.3 antibodies to test for binding and elimination of the malignant T-cell clone., Results: Therapeutic antibody candidates bound the malignant clone with high affinity. Antibodies killed engineered cell lines expressing the patient TCR Vβ13.3 by antibody-dependent cellular cytotoxicity and TCR-mediated activation-induced cell death, and exhibited specific killing of patient malignant T cells in combination with exogenous natural killer cells. EL4 cells expressing the patient's TCR Vβ13.3 were also killed by antibody administration in an in vivo murine model., Conclusions: This approach serves as an outline for development of therapeutics that can treat clonal T-cell-based malignancies and potentially other T-cell-mediated diseases. See related commentary by Varma and Diefenbach, p. 4024., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
8. Secondary fusion proteins as a mechanism of BCR::ABL1 kinase-independent resistance in chronic myeloid leukaemia.
- Author
-
Barnes EJ, Eide CA, Kaempf A, Bottomly D, Romine KA, Wilmot B, Saunders D, McWeeney SK, Tognon CE, and Druker BJ
- Subjects
- Humans, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Mutation, Cell Line, Drug Resistance, Neoplasm genetics, Fusion Proteins, bcr-abl metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism
- Abstract
Drug resistance in chronic myeloid leukaemia (CML) may occur via mutations in the causative BCR::ABL1 fusion or BCR::ABL1-independent mechanisms. We analysed 48 patients with BCR::ABL1-independent resistance for the presence of secondary fusion genes by RNA sequencing. We identified 10 of the most frequently detected secondary fusions in 21 patients. Validation studies, cell line models, gene expression analysis and drug screening revealed differences with respect to proliferation rate, differentiation and drug sensitivity. Notably, expression of RUNX1::MECOM led to resistance to ABL1 tyrosine kinase inhibitors in vitro. These results suggest secondary fusions contribute to BCR::ABL1-independent resistance and may be amenable to combined therapies., (© 2022 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
9. Loss of G0/G1 switch gene 2 (G0S2) promotes disease progression and drug resistance in chronic myeloid leukaemia (CML) by disrupting glycerophospholipid metabolism.
- Author
-
Gonzalez MA, Olivas IM, Bencomo-Alvarez AE, Rubio AJ, Barreto-Vargas C, Lopez JL, Dang SK, Solecki JP, McCall E, Astudillo G, Velazquez VV, Schenkel K, Reffell K, Perkins M, Nguyen N, Apaflo JN, Alvidrez E, Young JE, Lara JJ, Yan D, Senina A, Ahmann J, Varley KE, Mason CC, Eide CA, Druker BJ, Nurunnabi M, Padilla O, Bajpeyi S, and Eiring AM
- Subjects
- Animals, Mice, Disease Progression, Fusion Proteins, bcr-abl genetics, Genes, Switch, Protein Kinase Inhibitors therapeutic use, Humans, Drug Resistance, Neoplasm genetics, Glycerophospholipids metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Cell Cycle Proteins genetics
- Abstract
Tyrosine kinase inhibitors (TKIs) targeting BCR::ABL1 have turned chronic myeloid leukaemia (CML) from a fatal disease into a manageable condition for most patients. Despite improved survival, targeting drug-resistant leukaemia stem cells (LSCs) remains a challenge for curative CML therapy. Aberrant lipid metabolism can have a large impact on membrane dynamics, cell survival and therapeutic responses in cancer. While ceramide and sphingolipid levels were previously correlated with TKI response in CML, the role of lipid metabolism in TKI resistance is not well understood. We have identified downregulation of a critical regulator of lipid metabolism, G0/G1 switch gene 2 (G0S2), in multiple scenarios of TKI resistance, including (1) BCR::ABL1 kinase-independent TKI resistance, (2) progression of CML from the chronic to the blast phase of the disease, and (3) in CML versus normal myeloid progenitors. Accordingly, CML patients with low G0S2 expression levels had a worse overall survival. G0S2 downregulation in CML was not a result of promoter hypermethylation or BCR::ABL1 kinase activity, but was rather due to transcriptional repression by MYC. Using CML cell lines, patient samples and G0s2 knockout (G0s2
-/- ) mice, we demonstrate a tumour suppressor role for G0S2 in CML and TKI resistance. Our data suggest that reduced G0S2 protein expression in CML disrupts glycerophospholipid metabolism, correlating with a block of differentiation that renders CML cells resistant to therapy. Altogether, our data unravel a new role for G0S2 in regulating myeloid differentiation and TKI response in CML, and suggest that restoring G0S2 may have clinical utility., (© 2022 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.)- Published
- 2022
- Full Text
- View/download PDF
10. Comprehensive molecular characterization of a rare case of Philadelphia chromosome-positive acute myeloid leukemia.
- Author
-
Rosenberg MW, Savage SL, Eide CA, Reister Schultz A, Cook RJ, Press RD, Rempfer C, Eickelberg G, Wilmot B, McWeeney SK, Tyner JW, Druker BJ, and Tognon CE
- Subjects
- Humans, Philadelphia Chromosome, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl metabolism, Fusion Proteins, bcr-abl therapeutic use, Phosphatidylinositol 3-Kinases genetics, Translocation, Genetic, Leukemia, Myelogenous, Chronic, BCR-ABL Positive diagnosis, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive therapy, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
The Philadelphia chromosome (Ph) resulting from the t(9;22) translocation generates the oncogenic BCR::ABL1 fusion protein that is most commonly associated with chronic myeloid leukemia (CML) and Ph-positive (Ph+) acute lymphoblastic leukemia (ALL). There are also rare instances of patients (≤1%) with newly diagnosed acute myeloid leukemia (AML) that harbor this translocation (Paietta et al., Leukemia 12: 1881 [1998]; Keung et al., Leuk Res 28: 579 [2004]; Soupir et al., Am J Clin Pathol 127: 642 [2007]). AML with BCR::ABL has only recently been provisionally classified by the World Health Organization as a diagnostically distinct subtype of AML. Discernment from the extremely close differential diagnosis of myeloid blast crisis CML is challenging, largely relying on medical history rather than clinical characteristics (Arber et al., Blood 127: 2391 [2016]). To gain insight into the genomic features underlying the evolution of AML with BCR::ABL, we identified a patient presenting with a high-risk myelodysplastic syndrome that acquired a BCR::ABL alteration after a peripheral blood stem cell transplant. Serial samples were collected and analyzed using whole-exome sequencing, RNA-seq, and ex vivo functional drug screens. Persistent subclones were identified, both at diagnosis and at relapse, including an SF3B1 p.Lys700Glu mutation that later cooccurred with an NRAS p.Gly12Cys mutation. Functional ex vivo drug screening performed on primary patient cells suggested that combination therapies of ABL1 with RAS or PI3K pathway inhibitors could have augmented the patient's response throughout the course of disease. Together, our findings argue for the importance of genomic profiling and the potential value of ABL1 inhibitor-inclusive combination treatment strategies in patients with this rare disease., (© 2022 Rosenberg et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2022
- Full Text
- View/download PDF
11. Integrative analysis of drug response and clinical outcome in acute myeloid leukemia.
- Author
-
Bottomly D, Long N, Schultz AR, Kurtz SE, Tognon CE, Johnson K, Abel M, Agarwal A, Avaylon S, Benton E, Blucher A, Borate U, Braun TP, Brown J, Bryant J, Burke R, Carlos A, Chang BH, Cho HJ, Christy S, Coblentz C, Cohen AM, d'Almeida A, Cook R, Danilov A, Dao KT, Degnin M, Dibb J, Eide CA, English I, Hagler S, Harrelson H, Henson R, Ho H, Joshi SK, Junio B, Kaempf A, Kosaka Y, Laderas T, Lawhead M, Lee H, Leonard JT, Lin C, Lind EF, Liu SQ, Lo P, Loriaux MM, Luty S, Maxson JE, Macey T, Martinez J, Minnier J, Monteblanco A, Mori M, Morrow Q, Nelson D, Ramsdill J, Rofelty A, Rogers A, Romine KA, Ryabinin P, Saultz JN, Sampson DA, Savage SL, Schuff R, Searles R, Smith RL, Spurgeon SE, Sweeney T, Swords RT, Thapa A, Thiel-Klare K, Traer E, Wagner J, Wilmot B, Wolf J, Wu G, Yates A, Zhang H, Cogle CR, Collins RH, Deininger MW, Hourigan CS, Jordan CT, Lin TL, Martinez ME, Pallapati RR, Pollyea DA, Pomicter AD, Watts JM, Weir SJ, Druker BJ, McWeeney SK, and Tyner JW
- Subjects
- Cell Differentiation, Cohort Studies, Humans, Receptors, Cell Surface genetics, Transcriptome, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Acute myeloid leukemia (AML) is a cancer of myeloid-lineage cells with limited therapeutic options. We previously combined ex vivo drug sensitivity with genomic, transcriptomic, and clinical annotations for a large cohort of AML patients, which facilitated discovery of functional genomic correlates. Here, we present a dataset that has been harmonized with our initial report to yield a cumulative cohort of 805 patients (942 specimens). We show strong cross-cohort concordance and identify features of drug response. Further, deconvoluting transcriptomic data shows that drug sensitivity is governed broadly by AML cell differentiation state, sometimes conditionally affecting other correlates of response. Finally, modeling of clinical outcome reveals a single gene, PEAR1, to be among the strongest predictors of patient survival, especially for young patients. Collectively, this report expands a large functional genomic resource, offers avenues for mechanistic exploration and drug development, and reveals tools for predicting outcome in AML., Competing Interests: Declaration of interests C.E.T. receives research support from Notable Labs and serves as a scientific liaison for AstraZeneca. J.E.M. receives research funding from Gilead Pharmaceutical and serves on a scientific advisory board for Ionis Pharmaceuticals. M.W.D. serves on the advisory boards and/or as a consultant for Novartis, Incyte, and BMS and receives research funding from BMS and Gilead. C.S.H. receives research funding from Sellas. T.L.L. consults for Jazz Pharmaceuticals and receives research funding from Tolero, Gilead, Prescient, Ono, Bio-Path, Mateon, Genentech/Roche, Trovagene, AbbVie, Pfizer, Celgene, Imago, Astellas, Karyopharm, Seattle Genetics, and Incyte. D.A.P. receives research funding from Pfizer and Agios and served on advisory boards for Pfizer, Celyad, Agios, Celgene, AbbVie, Argenx, Takeda, and Servier. B.J.D. serves on the advisory boards for Aileron Therapeutics, Aptose, Blueprint Medicines, Cepheid, EnLiven Therapeutics, Gilead, GRAIL, Iterion Therapeutics, Nemucore Medical Innovations, the Novartis CML Molecular Monitoring Steering Committee, Recludix Pharma, the RUNX1 Research Program, ALLCRON Pharma, VB Therapeutics, Vincerx Pharma, and the Board of Directors for Amgen, and receives research funding from EnLiven and Recludix. B.J.D. is principal investigator or co-investigator on Novartis, BMS, and Pfizer clinical trials. His institution, OHSU, has contracts with these companies to pay for patient costs, nurse and data manager salaries, and institutional overhead, but he does not derive salary, nor does his laboratory receive funds, from these contracts. J.W.T. has received research support from Acerta, Agios, Aptose, Array, AstraZeneca, Constellation, Genentech, Gilead, Incyte, Janssen, Kronos, Meryx, Petra, Schrodinger, Seattle Genetics, Syros, Takeda, and Tolero and serves on the advisory board for Recludix Pharma. The authors certify that all compounds tested in this study were chosen without input from any of our industry partners. A subset of findings from this manuscript have been included in a pending patent application., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
12. Associating drug sensitivity with differentiation status identifies effective combinations for acute myeloid leukemia.
- Author
-
Kurtz SE, Eide CA, Kaempf A, Long N, Bottomly D, Nikolova O, Druker BJ, McWeeney SK, Chang BH, Tyner JW, and Agarwal A
- Subjects
- Cell Differentiation, Humans, Immunophenotyping, Proto-Oncogene Proteins c-bcl-2 metabolism, p38 Mitogen-Activated Protein Kinases, Leukemia, Myeloid, Acute genetics
- Abstract
Using ex vivo drug screening of primary patient specimens, we identified the combination of the p38 MAPK inhibitor doramapimod (DORA) with the BCL2 inhibitor venetoclax (VEN) as demonstrating broad, enhanced efficacy compared with each single agent across 335 acute myeloid leukemia (AML) patient samples while sparing primary stromal cells. Single-agent DORA and VEN sensitivity was associated with distinct, nonoverlapping tumor cell differentiation states. In particular, increased monocytes, M4/M5 French-American-British classification, and CD14+ immunophenotype tracked with sensitivity to DORA and resistance to VEN but were mitigated with the combination. Increased expression of MAPK14 and BCL2, the respective primary targets of DORA and VEN, were observed in monocytic and undifferentiated leukemias, respectively. Enrichment for DORA and VEN sensitivities was observed in AML with monocyte-like and progenitor-like transcriptomic signatures, respectively, and these associations diminished with the combination. The mechanism underlying the combination's enhanced efficacy may result from inhibition of p38 MAPK-mediated phosphorylation of BCL2, which in turn enhances sensitivity to VEN. These findings suggest exploiting complementary drug sensitivity profiles with respect to leukemic differentiation state, such as dual targeting of p38 MAPK and BCL2, offers opportunity for broad, enhanced efficacy across the clinically challenging heterogeneous landscape of AML., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
13. Genome-wide CRISPR screen identifies regulators of MAPK and MTOR pathways mediating sorafenib resistance in acute myeloid leukemia.
- Author
-
Damnernsawad A, Bottomly D, Kurtz SE, Eide CA, McWeeney SK, Tyner JW, and Nechiporuk T
- Subjects
- Cell Line, Tumor, Clustered Regularly Interspaced Short Palindromic Repeats, Humans, MAP Kinase Signaling System, Mutation, Niacinamide pharmacology, Niacinamide therapeutic use, Phenylurea Compounds pharmacology, Phenylurea Compounds therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, TOR Serine-Threonine Kinases genetics, Transcription Factors, fms-Like Tyrosine Kinase 3 genetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Sorafenib pharmacology
- Abstract
Drug resistance impedes the long-term effect of targeted therapies in acute myeloid leukemia (AML), necessitating the identification of mechanisms underlying resistance. Approximately 25% of AML patients carry FLT3 mutations and develop post-treatment insensitivity to FLT3 inhibitors, including sorafenib. Using a genome-wide CRISPR screen, we identified LZTR1, NF1, TSC1 or TSC2, negative regulators of the MAPK and MTOR pathways, as mediators of sorafenib resistance. Analyses of ex vivo drug sensitivity assays in FLT3-ITD AML patient samples revealed lower expression of LZTR1, NF1, and TSC2 correlated with sorafenib sensitivity. Importantly, MAPK and/or MTOR complex1 (MTORC1) activity were upregulated in AML cells made resistant to several FLT3 inhibitors, including crenolanib, quizartinib, or sorafenib. These cells were sensitive to MEK inhibitors, and the combination of FLT3 and MEK inhibitors showed enhanced efficacy, suggesting its effectiveness in AML patients with FLT3 mutations and those with resistance to FLT3 inhibitors.
- Published
- 2022
- Full Text
- View/download PDF
14. Aurora A kinase as a target for therapy in TCF3-HLF rearranged acute lymphoblastic leukemia.
- Author
-
Leonard J, Wolf JS, Degnin M, Eide CA, LaTocha D, Lenz K, Wilmot B, Mullighan CG, Loh M, Hunger SP, Druker BJ, Loriaux MM, Tyner JW, and Chang BH
- Subjects
- Basic Helix-Loop-Helix Transcription Factors, Humans, Oncogene Proteins, Fusion genetics, Translocation, Genetic, Aurora Kinase A genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2021
- Full Text
- View/download PDF
15. Lentiviral-Driven Discovery of Cancer Drug Resistance Mutations.
- Author
-
Yenerall P, Kollipara RK, Avila K, Peyton M, Eide CA, Bottomly D, McWeeney SK, Liu Y, Westover KD, Druker BJ, Minna JD, and Kittler R
- Subjects
- Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Cell Line, Tumor, Dose-Response Relationship, Drug, Gene Expression, Gene Expression Regulation, Neoplastic drug effects, Humans, Models, Molecular, Neoplasms drug therapy, Structure-Activity Relationship, Biomarkers, Tumor, Drug Discovery methods, Drug Resistance, Neoplasm genetics, Genetic Vectors genetics, Lentivirus genetics, Mutation, Neoplasms genetics
- Abstract
Identifying resistance mutations in a drug target provides crucial information. Lentiviral transduction creates multiple types of mutations due to the error-prone nature of the HIV-1 reverse transcriptase (RT). Here we optimized and leveraged this property to identify drug resistance mutations, developing a technique we term LentiMutate. This technique was validated by identifying clinically relevant EGFR resistance mutations, then applied to two additional clinical anticancer drugs: imatinib, a BCR-ABL inhibitor, and AMG 510, a KRAS G12C inhibitor. Novel deletions in BCR-ABL1 conferred resistance to imatinib. In KRAS-G12C or wild-type KRAS, point mutations in the AMG 510 binding pocket or oncogenic non-G12C mutations conferred resistance to AMG 510. LentiMutate should prove highly valuable for clinical and preclinical cancer-drug development. SIGNIFICANCE: LentiMutate can evaluate a drug's on-target activity and can nominate resistance mutations before they occur in patients, which could accelerate and refine drug development to increase the survival of patients with cancer., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
16. NT157, an IGF1R-IRS1/2 inhibitor, exhibits antineoplastic effects in pre-clinical models of chronic myeloid leukemia.
- Author
-
Scopim-Ribeiro R, Machado-Neto JA, Eide CA, Coelho-Silva JL, Fenerich BA, Fernandes JC, Scheucher PS, Savage Stevens SL, de Melo Campos P, Olalla Saad ST, de Carvalho Palma L, de Figueiredo-Pontes LL, Simões BP, Rego EM, Tognon CE, Druker BJ, and Traina F
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Disease Models, Animal, Drug Resistance, Neoplasm drug effects, Fusion Proteins, bcr-abl antagonists & inhibitors, Gene Expression Regulation, Neoplastic, Humans, Imatinib Mesylate pharmacology, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Mice, Protein Kinase Inhibitors pharmacology, Pyrogallol pharmacology, Pyrogallol therapeutic use, Sulfonamides pharmacology, Antineoplastic Agents therapeutic use, Insulin Receptor Substrate Proteins antagonists & inhibitors, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrogallol analogs & derivatives, Receptor, IGF Type 1 antagonists & inhibitors, Sulfonamides therapeutic use
- Abstract
Chronic myeloid leukemia (CML) is successfully treated with BCR-ABL1 tyrosine kinase inhibitors, but a significant percentage of patients develop resistance. Insulin receptor substrate 1 (IRS1) has been shown to constitutively associate with BCR-ABL1, and IRS1-specific silencing leads to antineoplastic effects in CML cell lines. Here, we characterized the efficacy of NT157, a pharmacological inhibitor of IGF1R-IRS1/2, in CML cells and observed significantly reduced cell viability and proliferation, accompanied by induction of apoptosis. In human K562 cells and in murine Ba/F3 cells, engineered to express either wild-type BCR-ABL1 or the imatinib-resistant BCR-ABL1
T315I mutant, NT157 inhibited BCR-ABL1, IGF1R, IRS1/2, PI3K/AKT/mTOR, and STAT3/5 signaling, increased CDKN1A, FOS and JUN tumor suppressor gene expression, and reduced MYC and BCL2 oncogenes. NT157 significantly reduced colony formation of human primary CML cells with minimal effect on normal hematopoietic cells. Exposure of primary CML cells harboring BCR-ABL1T315I to NT157 resulted in increased apoptosis, reduced cell proliferation and decreased phospho-CRKL levels. In conclusion, NT157 has antineoplastic effects on BCR-ABL1 leukemogenesis, independent of T315I mutational status.- Published
- 2021
- Full Text
- View/download PDF
17. Proteasome 26S subunit, non-ATPases 1 (PSMD1) and 3 (PSMD3), play an oncogenic role in chronic myeloid leukemia by stabilizing nuclear factor-kappa B.
- Author
-
Bencomo-Alvarez AE, Rubio AJ, Olivas IM, Gonzalez MA, Ellwood R, Fiol CR, Eide CA, Lara JJ, Barreto-Vargas C, Jave-Suarez LF, Nteliopoulos G, Reid AG, Milojkovic D, Druker BJ, Apperley J, Khorashad JS, and Eiring AM
- Subjects
- Animals, Apoptosis physiology, Drug Resistance, Neoplasm, Heterografts, Humans, K562 Cells, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Mice, Mice, Nude, NF-kappa B genetics, Proteasome Endopeptidase Complex genetics, Protein Kinase Inhibitors pharmacology, Transcription, Genetic, Up-Regulation, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, NF-kappa B metabolism, Proteasome Endopeptidase Complex metabolism
- Abstract
Tyrosine kinase inhibitors (TKIs) targeting BCR-ABL1 have revolutionized therapy for chronic myeloid leukemia (CML), paving the way for clinical development in other diseases. Despite success, targeting leukemic stem cells and overcoming drug resistance remain challenges for curative cancer therapy. To identify drivers of kinase-independent TKI resistance in CML, we performed genome-wide expression analyses on TKI-resistant versus sensitive CML cell lines, revealing a nuclear factor-kappa B (NF-κB) expression signature. Nucleocytoplasmic fractionation and luciferase reporter assays confirmed increased NF-κB activity in the nucleus of TKI-resistant versus sensitive CML cell lines and CD34
+ patient samples. Two genes that were upregulated in TKI-resistant CML cells were proteasome 26S subunit, non-ATPases 1 (PSMD1) and 3 (PSMD3), both members of the 19S regulatory complex in the 26S proteasome. PSMD1 and PSMD3 were also identified as survival-critical genes in a published small hairpin RNA library screen of TKI resistance. We observed markedly higher levels of PSMD1 and PSMD3 mRNA in CML patients who had progressed to the blast phase compared with the chronic phase of the disease. Knockdown of PSMD1 or PSMD3 protein correlated with reduced survival and increased apoptosis in CML cells, but not in normal cord blood CD34+ progenitors. Luciferase reporter assays and immunoblot analyses demonstrated that PSMD1 and PSMD3 promote NF-κB protein expression in CML, and that signal transducer and activator of transcription 3 (STAT3) further activates NF-κB in scenarios of TKI resistance. Our data identify NF-κB as a transcriptional driver in TKI resistance, and implicate PSMD1 and PSMD3 as plausible therapeutic targets worthy of future investigation in CML and possibly other malignancies.- Published
- 2021
- Full Text
- View/download PDF
18. ERBB2/HER2 mutations are transforming and therapeutically targetable in leukemia.
- Author
-
Joshi SK, Keck JM, Eide CA, Bottomly D, Traer E, Tyner JW, McWeeney SK, Tognon CE, and Druker BJ
- Subjects
- Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Dose-Response Relationship, Drug, Drug Resistance, Neoplasm genetics, Female, Humans, Leukemia drug therapy, Leukemia metabolism, Leukemia pathology, Molecular Targeted Therapy, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 metabolism, Treatment Outcome, Biomarkers, Tumor, Cell Transformation, Neoplastic genetics, Leukemia genetics, Mutation, Receptor, ErbB-2 genetics
- Published
- 2020
- Full Text
- View/download PDF
19. Simultaneous kinase inhibition with ibrutinib and BCL2 inhibition with venetoclax offers a therapeutic strategy for acute myeloid leukemia.
- Author
-
Eide CA, Kurtz SE, Kaempf A, Long N, Agarwal A, Tognon CE, Mori M, Druker BJ, Chang BH, Danilov AV, and Tyner JW
- Subjects
- Adenine analogs & derivatives, Animals, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Cell Line, Tumor, Humans, Mice, Piperidines, Sulfonamides pharmacology, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Leukemia, Myeloid, Acute drug therapy, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Pyrazoles therapeutic use, Pyrimidines therapeutic use, Sulfonamides therapeutic use
- Abstract
Acute myeloid leukemia (AML) results from the enhanced proliferation and impaired differentiation of hematopoietic stem and progenitor cells. Using an ex vivo functional screening assay, we identified that the combination of the BTK inhibitor ibrutinib and BCL2 inhibitor venetoclax (IBR + VEN), currently in clinical trials for chronic lymphocytic leukemia (CLL), demonstrated enhanced efficacy on primary AML patient specimens, AML cell lines, and in a mouse xenograft model of AML. Expanded analyses among a large cohort of hematologic malignancies (n = 651 patients) revealed that IBR + VEN sensitivity associated with selected genetic and phenotypic features in both CLL and AML specimens. Among AML samples, 11q23 MLL rearrangements were highly sensitive to IBR + VEN. Analysis of differentially expressed genes with respect to IBR + VEN sensitivity indicated pathways preferentially enriched in patient samples with reduced ex vivo sensitivity, including IL-10 signaling. These findings suggest that IBR + VEN may represent an effective therapeutic option for patients with AML.
- Published
- 2020
- Full Text
- View/download PDF
20. Response and Resistance to BCR-ABL1-Targeted Therapies.
- Author
-
Braun TP, Eide CA, and Druker BJ
- Subjects
- Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Drug Resistance, Neoplasm genetics, Fusion Proteins, bcr-abl antagonists & inhibitors, Fusion Proteins, bcr-abl genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Molecular Targeted Therapy, Protein Kinase Inhibitors therapeutic use
- Abstract
Chronic myeloid leukemia (CML), caused by constitutively active BCR-ABL1 fusion tyrosine kinase, has served as a paradigm for successful application of molecularly targeted cancer therapy. The development of the tyrosine kinase inhibitor (TKI) imatinib allows patients with CML to experience near-normal life expectancy. Specific point mutations that decrease drug binding affinity can produce TKI resistance, and second- and third-generation TKIs largely mitigate this problem. Some patients develop TKI resistance without known resistance mutations, with significant heterogeneity in the underlying mechanism, but this is relatively uncommon, with the majority of patients with chronic phase CML achieving long-term disease control. In contrast, responses to TKI treatment are short lived in advanced phases of the disease or in BCR-ABL1-positive acute lymphoblastic leukemia, with relapse driven by both BCR-ABL1 kinase-dependent and -independent mechanisms. Additionally, the frontline CML treatment with second-generation TKIs produces deeper molecular responses, driving disease burden below the detection limit for a greater number of patients. For patients with deep molecular responses, up to half have been able to discontinue therapy. Current efforts are focused on identifying therapeutic strategies to drive deeper molecular responses, enabling more patients to attempt TKI discontinuation., Competing Interests: Declaration of Interests B.J.D. potential competing interests––SAB: Aileron Therapeutics, Therapy Architects (ALLCRON), Cepheid, Vivid Biosciences, Celgene, RUNX1 Research Program, EnLiven Therapeutics, Gilead Sciences (inactive), Monojul (inactive); SAB & Stock: Aptose Biosciences, Blueprint Medicines, Iterion Therapeutics, Third Coast Therapeutics, GRAIL (SAB inactive); Scientific Founder: MolecularMD (inactive, acquired by ICON); Board of Directors & Stock: Amgen; Board of Directors: Burroughs Wellcome Fund, CureOne; Joint Steering Committee: Beat AML LLS; Founder: VB Therapeutics; Clinical Trial Funding: Novartis, Bristol-Myers Squibb, Pfizer; Royalties from Patent 6958335 (Novartis exclusive license) and OHSU and Dana-Farber Cancer Institute (one Merck exclusive license). The remaining authors have no competing interests to declare., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
21. NT157 has antineoplastic effects and inhibits IRS1/2 and STAT3/5 in JAK2 V617F -positive myeloproliferative neoplasm cells.
- Author
-
Fenerich BA, Fernandes JC, Rodrigues Alves APN, Coelho-Silva JL, Scopim-Ribeiro R, Scheucher PS, Eide CA, Tognon CE, Druker BJ, Rego EM, Machado-Neto JA, and Traina F
- Subjects
- Apoptosis drug effects, Cell Cycle Checkpoints drug effects, Cell Cycle Checkpoints genetics, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Insulin Receptor Substrate Proteins antagonists & inhibitors, Insulin Receptor Substrate Proteins genetics, Myeloproliferative Disorders genetics, Myeloproliferative Disorders pathology, Polycythemia Vera genetics, Polycythemia Vera pathology, Pyrogallol pharmacology, STAT3 Transcription Factor antagonists & inhibitors, STAT3 Transcription Factor genetics, STAT5 Transcription Factor antagonists & inhibitors, STAT5 Transcription Factor genetics, Janus Kinase 2 genetics, Myeloproliferative Disorders drug therapy, Polycythemia Vera drug therapy, Pyrogallol analogs & derivatives, Sulfonamides pharmacology
- Abstract
Recent data indicate that IGF1R/IRS signaling is a potential therapeutic target in BCR-ABL1-negative myeloproliferative neoplasms (MPN); in this pathway, IRS2 is involved in the malignant transformation induced by JAK2
V617F , and upregulation of IGF1R signaling induces the MPN phenotype. NT157, a synthetic compound designed as an IGF1R-IRS1/2 inhibitor, has been shown to induce antineoplastic effects in solid tumors. Herein, we aimed to characterize the molecular and cellular effects of NT157 in JAK2V617F -positive MPN cell lines (HEL and SET2) and primary patient hematopoietic cells. In JAK2V617F cell lines, NT157 decreased cell viability, clonogenicity, and cell proliferation, resulting in increases in apoptosis and cell cycle arrest in the G2 /M phase (p < 0.05). NT157 treatment inhibited IRS1/2, JAK2/STAT, and NFκB signaling, and it activated the AP-1 complex, downregulated four oncogenes (CCND1, MYB, WT1, and NFKB1), and upregulated three apoptotic-related genes (CDKN1A, FOS, and JUN) (p < 0.05). NT157 induced genotoxic stress in a JAK2/STAT-independent manner. NT157 inhibited erythropoietin-independent colony formation in cells from polycythemia vera patients (p < 0.05). These findings further elucidate the mechanism of NT157 action in a MPN context and suggest that targeting IRS1/2 proteins may represent a promising therapeutic strategy for MPN.- Published
- 2020
- Full Text
- View/download PDF
22. Differentiation of leukemic blasts is not completely blocked in acute myeloid leukemia.
- Author
-
Agarwal A, Bolosky WJ, Wilson DB, Eide CA, Olson SB, Fan G, and Druker BJ
- Subjects
- Adolescent, Adult, Aged, Cell Death, Female, Gene Expression Regulation, Leukemic, Hematopoiesis, Hematopoietic Stem Cells pathology, Humans, Male, Middle Aged, Myeloid Cells pathology, Transcription Factors genetics, Blast Crisis pathology, Cell Differentiation physiology, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Models, Biological
- Abstract
Hematopoiesis, the formation of blood cells, involves the hierarchical differentiation of immature blast cells into mature, functional cell types and lineages of the immune system. Hematopoietic stem cells precisely regulate self-renewal versus differentiation to balance the production of blood cells and maintenance of the stem cell pool. The canonical view of acute myeloid leukemia (AML) is that it results from a combination of molecular events in a hematopoietic stem cell that block differentiation and drive proliferation. These events result in the accumulation of primitive hematopoietic blast cells in the blood and bone marrow. We used mathematical modeling to determine the impact of varying differentiation rates on myeloblastic accumulation. Our model shows that, instead of the commonly held belief that AML results from a complete block of differentiation of the hematopoietic stem cell, even a slight skewing of the fraction of cells that differentiate would produce an accumulation of blasts. We confirmed this model by interphase fluorescent in situ hybridization (FISH) and sequencing of purified cell populations from patients with AML, which showed that different leukemia-causing molecular abnormalities typically thought to block differentiation were consistently present in mature myeloid cells such as neutrophils and monocytes at similar levels to those in immature myeloid cells. These findings suggest reduced or skewed, rather than blocked, differentiation is responsible for the development of AML. Approaches that restore normal regulation of hematopoiesis could be effective treatment strategies., Competing Interests: Competing interest statement: B.J.D. potential competing interests: The Scientific Advisory Board (SAB): Aileron Therapeutics, ALLCRON, Cepheid, Vivid Biosciences, Celgene, RUNX1 Research Program, EnLiven Therapeutics, Gilead Sciences (inactive), Baxalta (inactive), and Monojul (inactive); SAB & Stock: Aptose Biosciences, Blueprint Medicines, Beta Cat, Third Coast Therapeutics, GRAIL (inactive), and CTI BioPharma (inactive); Scientific Founder: MolecularMD (inactive, acquired by ICON); Board of Directors & Stock: Amgen; Board of Directors: Burroughs Wellcome Fund, CureOne; Joint Steering Committee: Beat AML LLS; Founder: VP Therapeutics; Clinical Trial Funding: Novartis, Bristol-Myers Squibb, Pfizer; Royalties from Patent 6958335 (Novartis exclusive license) and Oregon Health & Science University (OHSU), and Dana-Farber Cancer Institute (one Merck exclusive license).
- Published
- 2019
- Full Text
- View/download PDF
23. Combining the Allosteric Inhibitor Asciminib with Ponatinib Suppresses Emergence of and Restores Efficacy against Highly Resistant BCR-ABL1 Mutants.
- Author
-
Eide CA, Zabriskie MS, Savage Stevens SL, Antelope O, Vellore NA, Than H, Schultz AR, Clair P, Bowler AD, Pomicter AD, Yan D, Senina AV, Qiang W, Kelley TW, Szankasi P, Heinrich MC, Tyner JW, Rea D, Cayuela JM, Kim DW, Tognon CE, O'Hare T, Druker BJ, and Deininger MW
- Subjects
- Allosteric Regulation drug effects, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Binding Sites drug effects, Binding Sites genetics, Cell Line, Tumor transplantation, Disease Models, Animal, Drug Resistance, Neoplasm genetics, Female, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl metabolism, Humans, Imidazoles therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Mice, Molecular Docking Simulation, Molecular Dynamics Simulation, Molecular Targeted Therapy methods, Mutation, Niacinamide pharmacology, Niacinamide therapeutic use, Primary Cell Culture, Pyrazoles therapeutic use, Pyridazines therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Drug Resistance, Neoplasm drug effects, Fusion Proteins, bcr-abl antagonists & inhibitors, Imidazoles pharmacology, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Niacinamide analogs & derivatives, Pyrazoles pharmacology, Pyridazines pharmacology
- Abstract
BCR-ABL1 point mutation-mediated resistance to tyrosine kinase inhibitor (TKI) therapy in Philadelphia chromosome-positive (Ph
+ ) leukemia is effectively managed with several approved drugs, including ponatinib for BCR-ABL1T315I -mutant disease. However, therapy options are limited for patients with leukemic clones bearing multiple BCR-ABL1 mutations. Asciminib, an allosteric inhibitor targeting the myristoyl-binding pocket of BCR-ABL1, is active against most single mutants but ineffective against all tested compound mutants. We demonstrate that combining asciminib with ATP site TKIs enhances target inhibition and suppression of resistant outgrowth in Ph+ clinical isolates and cell lines. Inclusion of asciminib restores ponatinib's effectiveness against currently untreatable compound mutants at clinically achievable concentrations. Our findings support combining asciminib with ponatinib as a treatment strategy for this molecularly defined group of patients., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
24. Targeting BCR-ABL1 in Chronic Myeloid Leukemia by PROTAC-Mediated Targeted Protein Degradation.
- Author
-
Burslem GM, Schultz AR, Bondeson DP, Eide CA, Savage Stevens SL, Druker BJ, and Crews CM
- Subjects
- Animals, Apoptosis drug effects, Cell Proliferation drug effects, Drug Resistance, Neoplasm, Humans, K562 Cells, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Mice, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Protein Array Analysis, Tumor Cells, Cultured, Fusion Proteins, bcr-abl antagonists & inhibitors, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Neoplastic Stem Cells drug effects, Protein Kinase Inhibitors pharmacology, Proteolysis drug effects
- Abstract
Although the use of ATP-competitive tyrosine kinase inhibitors of oncoprotein BCR-ABL1 has enabled durable responses in patients with chronic myeloid leukemia (CML), issues of drug resistance and residual leukemic stem cells remain. To test whether the degradation of BCR-ABL1 kinase could offer improved response, we developed a series of proteolysis-targeting chimera (PROTAC) that allosterically target BCR-ABL1 protein and recruit the E3 ligase Von Hippel-Lindau, resulting in ubiquitination and subsequent degradation of the oncogenic fusion protein. In both human CML K562 cells and murine Ba/F3 cells expressing BCR-ABL1, lead compound GMB-475 induced rapid proteasomal degradation and inhibition of downstream biomarkers, such as STAT5, and showed increased sensitivity compared with diastereomeric controls lacking degradation activity. Notably, GMB-475 inhibited the proliferation of certain clinically relevant BCR-ABL1 kinase domain point mutants and further sensitized Ba/F3 BCR-ABL1 cells to inhibition by imatinib, while demonstrating no toxicity toward Ba/F3 parental cells. Reverse phase protein array analysis suggested additional differences in levels of phosphorylated SHP2, GAB2, and SHC associated with BCR-ABL1 degradation. Importantly, GMB-475 reduced viability and increased apoptosis in primary CML CD34
+ cells, with no effect on healthy CD34+ cells at identical concentrations. GMB-475 degraded BCR-ABL1 and reduced cell viability in primary CML stem cells. Together, these findings suggest that combined BCR-ABL1 kinase inhibition and protein degradation may represent a strategy to address BCR-ABL1-dependent drug resistance, and warrant further investigation into the eradication of persistent leukemic stem cells, which rely on neither the presence nor the activity of the BCR-ABL1 protein for survival. SIGNIFICANCE: Small-molecule-induced degradation of BCR-ABL1 in CML provides an advantage over inhibition and provides insights into CML stem cell biology. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/18/4744/F1.large.jpg., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
25. Genomic landscape of neutrophilic leukemias of ambiguous diagnosis.
- Author
-
Zhang H, Wilmot B, Bottomly D, Dao KT, Stevens E, Eide CA, Khanna V, Rofelty A, Savage S, Reister Schultz A, Long N, White L, Carlos A, Henson R, Lin C, Searles R, Collins RH, DeAngelo DJ, Deininger MW, Dunn T, Hein T, Luskin MR, Medeiros BC, Oh ST, Pollyea DA, Steensma DP, Stone RM, Druker BJ, McWeeney SK, Maxson JE, Gotlib JR, and Tyner JW
- Subjects
- Adult, Aged, Aged, 80 and over, Cells, Cultured, Cohort Studies, DNA Mutational Analysis, Diagnosis, Differential, Female, Gene Expression Profiling, Genomics, HEK293 Cells, Humans, Male, Middle Aged, Mutation, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes genetics, Prognosis, Hematologic Neoplasms diagnosis, Hematologic Neoplasms genetics, Leukemia, Neutrophilic, Chronic diagnosis, Leukemia, Neutrophilic, Chronic genetics
- Abstract
Chronic neutrophilic leukemia (CNL), atypical chronic myeloid leukemia (aCML), and myelodysplastic/myeloproliferative neoplasms, unclassifiable (MDS/MPN-U) are a group of rare and heterogeneous myeloid disorders. There is strong morphologic resemblance among these distinct diagnostic entities as well as a lack of specific molecular markers and limited understanding of disease pathogenesis, which has made diagnosis challenging in certain cases. The treatment has remained empirical, resulting in dismal outcomes. We, therefore, performed whole-exome and RNA sequencing of these rare hematologic malignancies and present the most complete survey of the genomic landscape of these diseases to date. We observed a diversity of combinatorial mutational patterns that generally do not cluster within any one diagnosis. Gene expression analysis reveals enrichment, but not cosegregation, of clinical and genetic disease features with transcriptional clusters. In conclusion, these groups of diseases represent a continuum of related diseases rather than discrete diagnostic entities., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
26. Laying the foundation for genomically-based risk assessment in chronic myeloid leukemia.
- Author
-
Branford S, Kim DDH, Apperley JF, Eide CA, Mustjoki S, Ong ST, Nteliopoulos G, Ernst T, Chuah C, Gambacorti-Passerini C, Mauro MJ, Druker BJ, Kim DW, Mahon FX, Cortes J, Radich JP, Hochhaus A, and Hughes TP
- Subjects
- Genes, Neoplasm, Hematopoiesis, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive etiology, Mutation, Protein Kinase Inhibitors therapeutic use, Protein-Tyrosine Kinases antagonists & inhibitors, Repressor Proteins genetics, Risk Assessment, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics
- Abstract
Outcomes for patients with chronic myeloid leukemia (CML) have substantially improved due to advances in drug development and rational treatment intervention strategies. Despite these significant advances there are still unanswered questions on patient management regarding how to more reliably predict treatment failure at the time of diagnosis and how to select frontline tyrosine kinase inhibitor (TKI) therapy for optimal outcome. The BCR-ABL1 transcript level at diagnosis has no established prognostic impact and cannot guide frontline TKI selection. BCR-ABL1 mutations are detected in ~50% of TKI resistant patients but are rarely responsible for primary resistance. Other resistance mechanisms are largely uncharacterized and there are no other routine molecular testing strategies to facilitate the evaluation and further stratification of TKI resistance. Advances in next-generation sequencing technology has aided the management of a growing number of other malignancies, enabling the incorporation of somatic mutation profiles in diagnosis, classification, and prognostication. A largely unexplored area in CML research is whether expanded genomic analysis at diagnosis, resistance, and disease transformation can enhance patient management decisions, as has occurred for other cancers. The aim of this article is to review publications that reported mutated cancer-associated genes in CML patients at various disease phases. We discuss the frequency and type of such variants at initial diagnosis and at the time of treatment failure and transformation. Current limitations in the evaluation of mutants and recommendations for future reporting are outlined. The collective evaluation of mutational studies over more than a decade suggests a limited set of cancer-associated genes are indeed recurrently mutated in CML and some at a relatively high frequency. Genomic studies have the potential to lay the foundation for improved diagnostic risk classification according to clinical and genomic risk, and to enable more precise early identification of TKI resistance.
- Published
- 2019
- Full Text
- View/download PDF
27. Clinical resistance to crenolanib in acute myeloid leukemia due to diverse molecular mechanisms.
- Author
-
Zhang H, Savage S, Schultz AR, Bottomly D, White L, Segerdell E, Wilmot B, McWeeney SK, Eide CA, Nechiporuk T, Carlos A, Henson R, Lin C, Searles R, Ho H, Lam YL, Sweat R, Follit C, Jain V, Lind E, Borthakur G, Garcia-Manero G, Ravandi F, Kantarjian HM, Cortes J, Collins R, Buelow DR, Baker SD, Druker BJ, and Tyner JW
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Benzimidazoles therapeutic use, Cell Line, Tumor, Drug Screening Assays, Antitumor, Epigenesis, Genetic drug effects, Female, GTP Phosphohydrolases genetics, HEK293 Cells, Humans, Inhibitory Concentration 50, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Male, Membrane Proteins genetics, Mice, Middle Aged, Mutation drug effects, Mutation genetics, Piperidines therapeutic use, Protein Kinase Inhibitors therapeutic use, Pyridones pharmacology, Pyridones therapeutic use, Pyrimidinones pharmacology, Pyrimidinones therapeutic use, Tandem Repeat Sequences genetics, Treatment Outcome, Exome Sequencing, Antineoplastic Combined Chemotherapy Protocols pharmacology, Benzimidazoles pharmacology, Drug Resistance, Neoplasm genetics, Leukemia, Myeloid, Acute drug therapy, Piperidines pharmacology, Protein Kinase Inhibitors pharmacology, fms-Like Tyrosine Kinase 3 genetics
- Abstract
FLT3 mutations are prevalent in AML patients and confer poor prognosis. Crenolanib, a potent type I pan-FLT3 inhibitor, is effective against both internal tandem duplications and resistance-conferring tyrosine kinase domain mutations. While crenolanib monotherapy has demonstrated clinical benefit in heavily pretreated relapsed/refractory AML patients, responses are transient and relapse eventually occurs. Here, to investigate the mechanisms of crenolanib resistance, we perform whole exome sequencing of AML patient samples before and after crenolanib treatment. Unlike other FLT3 inhibitors, crenolanib does not induce FLT3 secondary mutations, and mutations of the FLT3 gatekeeper residue are infrequent. Instead, mutations of NRAS and IDH2 arise, mostly as FLT3-independent subclones, while TET2 and IDH1 predominantly co-occur with FLT3-mutant clones and are enriched in crenolanib poor-responders. The remaining patients exhibit post-crenolanib expansion of mutations associated with epigenetic regulators, transcription factors, and cohesion factors, suggesting diverse genetic/epigenetic mechanisms of crenolanib resistance. Drug combinations in experimental models restore crenolanib sensitivity.
- Published
- 2019
- Full Text
- View/download PDF
28. Functional genomic landscape of acute myeloid leukaemia.
- Author
-
Tyner JW, Tognon CE, Bottomly D, Wilmot B, Kurtz SE, Savage SL, Long N, Schultz AR, Traer E, Abel M, Agarwal A, Blucher A, Borate U, Bryant J, Burke R, Carlos A, Carpenter R, Carroll J, Chang BH, Coblentz C, d'Almeida A, Cook R, Danilov A, Dao KT, Degnin M, Devine D, Dibb J, Edwards DK 5th, Eide CA, English I, Glover J, Henson R, Ho H, Jemal A, Johnson K, Johnson R, Junio B, Kaempf A, Leonard J, Lin C, Liu SQ, Lo P, Loriaux MM, Luty S, Macey T, MacManiman J, Martinez J, Mori M, Nelson D, Nichols C, Peters J, Ramsdill J, Rofelty A, Schuff R, Searles R, Segerdell E, Smith RL, Spurgeon SE, Sweeney T, Thapa A, Visser C, Wagner J, Watanabe-Smith K, Werth K, Wolf J, White L, Yates A, Zhang H, Cogle CR, Collins RH, Connolly DC, Deininger MW, Drusbosky L, Hourigan CS, Jordan CT, Kropf P, Lin TL, Martinez ME, Medeiros BC, Pallapati RR, Pollyea DA, Swords RT, Watts JM, Weir SJ, Wiest DL, Winters RM, McWeeney SK, and Druker BJ
- Subjects
- Core Binding Factor Alpha 2 Subunit genetics, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methyltransferase 3A, Datasets as Topic, Exome genetics, Female, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism, Male, Molecular Targeted Therapy, Nuclear Proteins genetics, Nucleophosmin, Proto-Oncogene Proteins genetics, Repressor Proteins genetics, Sequence Analysis, RNA, Serine-Arginine Splicing Factors genetics, Gene Expression Regulation, Neoplastic genetics, Genome, Human genetics, Genomics, Leukemia, Myeloid, Acute genetics
- Abstract
The implementation of targeted therapies for acute myeloid leukaemia (AML) has been challenging because of the complex mutational patterns within and across patients as well as a dearth of pharmacologic agents for most mutational events. Here we report initial findings from the Beat AML programme on a cohort of 672 tumour specimens collected from 562 patients. We assessed these specimens using whole-exome sequencing, RNA sequencing and analyses of ex vivo drug sensitivity. Our data reveal mutational events that have not previously been detected in AML. We show that the response to drugs is associated with mutational status, including instances of drug sensitivity that are specific to combinatorial mutational events. Integration with RNA sequencing also revealed gene expression signatures, which predict a role for specific gene networks in the drug response. Collectively, we have generated a dataset-accessible through the Beat AML data viewer (Vizome)-that can be leveraged to address clinical, genomic, transcriptomic and functional analyses of the biology of AML.
- Published
- 2018
- Full Text
- View/download PDF
29. Dual inhibition of JAK1/2 kinases and BCL2: a promising therapeutic strategy for acute myeloid leukemia.
- Author
-
Kurtz SE, Eide CA, Kaempf A, Mori M, Tognon CE, Borate U, Druker BJ, and Tyner JW
- Subjects
- Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols, Biomarkers, Tumor, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical methods, Drug Resistance, Neoplasm genetics, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Molecular Targeted Therapy, Mutation, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Janus Kinase 1 antagonists & inhibitors, Janus Kinase 2 antagonists & inhibitors, Leukemia, Myeloid, Acute metabolism, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors
- Published
- 2018
- Full Text
- View/download PDF
30. Synthetic lethality of TNK2 inhibition in PTPN11-mutant leukemia.
- Author
-
Jenkins C, Luty SB, Maxson JE, Eide CA, Abel ML, Togiai C, Nemecek ER, Bottomly D, McWeeney SK, Wilmot B, Loriaux M, Chang BH, and Tyner JW
- Subjects
- Animals, Child, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute enzymology, Leukemia, Myeloid, Acute genetics, Leukemia, Myelomonocytic, Juvenile drug therapy, Leukemia, Myelomonocytic, Juvenile enzymology, Leukemia, Myelomonocytic, Juvenile genetics, Male, Mice, Prognosis, Protein Kinase Inhibitors pharmacology, Signal Transduction, Survival Rate, Tumor Stem Cell Assay, Dasatinib pharmacology, Leukemia, Myeloid, Acute pathology, Leukemia, Myelomonocytic, Juvenile pathology, Protein Tyrosine Phosphatase, Non-Receptor Type 11 genetics, Protein-Tyrosine Kinases antagonists & inhibitors, Synthetic Lethal Mutations
- Abstract
The protein tyrosine phosphatase PTPN11 is implicated in the pathogenesis of juvenile myelomonocytic leukemia (JMML), acute myeloid leukemia (AML), and other malignancies. Activating mutations in PTPN11 increase downstream proliferative signaling and cell survival. We investigated the signaling upstream of PTPN11 in JMML and AML cells and found that PTPN11 was activated by the nonreceptor tyrosine/serine/threonine kinase TNK2 and that PTPN11-mutant JMML and AML cells were sensitive to TNK2 inhibition. In cultured human cell-based assays, PTPN11 and TNK2 interacted directly, enabling TNK2 to phosphorylate PTPN11, which subsequently dephosphorylated TNK2 in a negative feedback loop. Mutations in PTPN11 did not affect this physical interaction but increased the basal activity of PTPN11 such that TNK2-mediated activation was additive. Consequently, coexpression of TNK2 and mutant PTPN11 synergistically increased mitogen-activated protein kinase (MAPK) signaling and enhanced colony formation in bone marrow cells from mice. Chemical inhibition of TNK2 blocked MAPK signaling and colony formation in vitro and decreased disease burden in a patient with PTPN11-mutant JMML who was treated with the multikinase (including TNK2) inhibitor dasatinib. Together, these data suggest that TNK2 is a promising therapeutic target for PTPN11-mutant leukemias., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
31. Targeting of colony-stimulating factor 1 receptor (CSF1R) in the CLL microenvironment yields antineoplastic activity in primary patient samples.
- Author
-
Edwards V DK, Sweeney DT, Ho H, Eide CA, Rofelty A, Agarwal A, Liu SQ, Danilov AV, Lee P, Chantry D, McWeeney SK, Druker BJ, Tyner JW, Spurgeon SE, and Loriaux MM
- Abstract
In many malignancies, the tumor microenvironment includes CSF1R-expressing supportive monocyte/macrophages that promote tumor cell survival. For chronic lymphocytic leukemia (CLL), these supportive monocyte/macrophages are known as nurse-like cells (NLCs), although the potential effectiveness of selective small-molecule inhibitors of CSF1R against CLL is understudied. Here, we demonstrate the preclinical activity of two inhibitors of CSF1R, GW-2580 and ARRY-382, in primary CLL patient samples. We observed at least 25% of CLL samples showed sub-micromolar sensitivity to CSF1R inhibitors. This sensitivity was observed in samples with varying genetic and clinical backgrounds, although higher white cell count and monocyte cell percentage was associated with increased sensitivity. Depleting CD14-expressing monocytes preferentially decreased viability in samples sensitive to CSF1R inhibitors, and treating samples with CSF1R inhibitors eliminated the presence of NLCs in long-term culture conditions. These results indicate that CSF1R small-molecule inhibitors target CD14-expressing monocytes in the CLL microenvironment, thereby depriving leukemia cells of extrinsic support signals. In addition, significant synergy was observed combining CSF1R inhibitors with idelalisib or ibrutinib, two current CLL therapies that disrupt tumor cell intrinsic B-cell receptor signaling. These findings support the concept of simultaneously targeting supportive NLCs and CLL cells and demonstrate the potential clinical utility of this combination., Competing Interests: CONFLICTS OF INTEREST J.W.T. receives research support from Agios Pharmaceuticals, Array Biopharma, Aptose Biosciences, AstraZeneca, Constellation Pharmaceuticals, Genentech, Incyte, Janssen Research & Development, Seattle Genetics, and Takeda Pharmaceuticals, and is a consultant for Leap Oncology. S.E.S. receives research support from Bristol-Myers Squibb, Genentech, Janssen, Gilead, and Acerta and has received an honorarium from Gilead. AA receives research funding from CTI BioPharma. PL and DC are employed at Array Biopharma. A.V.D. Millenium Pharmaceuticals: Research Funding; Gilead Sciences Inc.: Research Funding; B.J.D.: Fred Hutchinson Cancer Research Center: Research Funding; Bristol-Myers Squibb: Research Funding; Henry Stewart Talks: Patents & Royalties; Millipore: Patents & Royalties; Sage Bionetworks: Research Funding; MolecularMD: Consultancy, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees; Gilead Sciences: Consultancy, Membership on an entity’s Board of Directors or advisory committees; Cylene Pharmaceuticals: Consultancy, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees; AstraZeneca: Consultancy; Novartis Pharmaceuticals: Research Funding; Blueprint Medicines: Consultancy, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees; Oregon Health & Science University: Patents & Royalties; CTI Biosciences: Consultancy, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees; Leukemia & Lymphoma Society: Membership on an entity’s Board of Directors or advisory committees, Research Funding; Oncotide Pharmaceuticals: Research Funding ; Roche TCRC, Inc.: Consultancy, Membership on an entity’s Board of Directors or advisory committees; McGraw Hill: Patents & Royalties; ARIAD: Research Funding; Aptose Therapeutics, Inc (formerly Lorus): Consultancy, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees. All other researchers have no relevant conflicts to disclose.
- Published
- 2018
- Full Text
- View/download PDF
32. Metformin exerts multitarget antileukemia activity in JAK2 V617F -positive myeloproliferative neoplasms.
- Author
-
Machado-Neto JA, Fenerich BA, Scopim-Ribeiro R, Eide CA, Coelho-Silva JL, Dechandt CRP, Fernandes JC, Rodrigues Alves APN, Scheucher PS, Simões BP, Alberici LC, de Figueiredo Pontes LL, Tognon CE, Druker BJ, Rego EM, and Traina F
- Subjects
- Animals, Cell Cycle drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Cyclin D1 genetics, Cyclin D1 metabolism, Female, Gene Knock-In Techniques, Humans, Janus Kinase 2 genetics, Mice, Mice, Inbred NOD, Mutation, Missense, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders physiopathology, Phosphorylation drug effects, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, STAT5 Transcription Factor genetics, STAT5 Transcription Factor metabolism, Antineoplastic Agents administration & dosage, Janus Kinase 2 metabolism, Metformin administration & dosage, Myeloproliferative Disorders drug therapy
- Abstract
The recurrent gain-of-function JAK2
V617F mutation confers growth factor-independent proliferation for hematopoietic cells and is a major contributor to the pathogenesis of myeloproliferative neoplasms (MPN). The lack of complete response in most patients treated with the JAK1/2 inhibitor ruxolitinib indicates the need for identifying novel therapeutic strategies. Metformin is a biguanide that exerts selective antineoplastic activity in hematological malignancies. In the present study, we investigate and compare effects of metformin and ruxolitinib alone and in combination on cell signaling and cellular functions in JAK2V617F -positive cells. In JAK2V617F -expressing cell lines, metformin treatment significantly reduced cell viability, cell proliferation, clonogenicity, and cellular oxygen consumption and delayed cell cycle progression. Metformin reduced cyclin D1 expression and RB, STAT3, STAT5, ERK1/2 and p70S6K phosphorylation. Metformin plus ruxolitinib demonstrated more intense reduction of cell viability and induction of apoptosis compared to monotherapy. Notably, metformin reduced Ba/F3 JAK2V617F tumor burden and splenomegaly in Jak2V617F knock-in-induced MPN mice and spontaneous erythroid colony formation in primary cells from polycythemia vera patients. In conclusion, metformin exerts multitarget antileukemia activity in MPN: downregulation of JAK2/STAT signaling and mitochondrial activity. Our exploratory study establishes novel molecular mechanisms of metformin and ruxolitinib action and provides insights for development of alternative/complementary therapeutic strategies for MPN.- Published
- 2018
- Full Text
- View/download PDF
33. A novel AGGF1-PDGFRb fusion in pediatric T-cell acute lymphoblastic leukemia.
- Author
-
Zabriskie MS, Antelope O, Verma AR, Draper LR, Eide CA, Pomicter AD, Tran TH, Druker BJ, Tyner JW, Miles RR, Graham JM, Hwang JY, Varley KE, Toydemir RM, Deininger MW, Raetz EA, and O'Hare T
- Subjects
- Child, Preschool, Fatal Outcome, Gene Rearrangement, Humans, Male, Protein Kinase Inhibitors therapeutic use, Angiogenic Proteins genetics, Oncogene Proteins, Fusion genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Receptor, Platelet-Derived Growth Factor beta genetics
- Published
- 2018
- Full Text
- View/download PDF
34. CRISPR-Cas9-mediated saturated mutagenesis screen predicts clinical drug resistance with improved accuracy.
- Author
-
Ma L, Boucher JI, Paulsen J, Matuszewski S, Eide CA, Ou J, Eickelberg G, Press RD, Zhu LJ, Druker BJ, Branford S, Wolfe SA, Jensen JD, Schiffer CA, Green MR, and Bolon DN
- Subjects
- Animals, Antineoplastic Agents pharmacology, CRISPR-Cas Systems drug effects, Cell Line, Tumor, Clustered Regularly Interspaced Short Palindromic Repeats drug effects, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Drug Resistance, Neoplasm drug effects, Fusion Proteins, bcr-abl genetics, Leukemia drug therapy, Leukemia genetics, Mice, Mutagenesis drug effects, Oncogenes genetics, Point Mutation drug effects, Point Mutation genetics, CRISPR-Cas Systems genetics, Drug Resistance, Neoplasm genetics, Mutagenesis genetics
- Abstract
Developing tools to accurately predict the clinical prevalence of drug-resistant mutations is a key step toward generating more effective therapeutics. Here we describe a high-throughput CRISPR-Cas9-based saturated mutagenesis approach to generate comprehensive libraries of point mutations at a defined genomic location and systematically study their effect on cell growth. As proof of concept, we mutagenized a selected region within the leukemic oncogene BCR-ABL1 Using bulk competitions with a deep-sequencing readout, we analyzed hundreds of mutations under multiple drug conditions and found that the effects of mutations on growth in the presence or absence of drug were critical for predicting clinically relevant resistant mutations, many of which were cancer adaptive in the absence of drug pressure. Using this approach, we identified all clinically isolated BCR-ABL1 mutations and achieved a prediction score that correlated highly with their clinical prevalence. The strategy described here can be broadly applied to a variety of oncogenes to predict patient mutations and evaluate resistance susceptibility in the development of new therapeutics., Competing Interests: The authors declare no conflict of interest., (Published under the PNAS license.)
- Published
- 2017
- Full Text
- View/download PDF
35. Functional validation of the oncogenic cooperativity and targeting potential of tuberous sclerosis mutation in medulloblastoma using a MYC-amplified model cell line.
- Author
-
Henderson JJ, Wagner JP, Hofmann NE, Eide CA, Cho YJ, Druker BJ, and Davare MA
- Subjects
- Amino Acid Substitution, Cell Line, Tumor, Humans, Medulloblastoma metabolism, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism, Proteolysis, Proto-Oncogene Proteins c-myc metabolism, Tuberous Sclerosis Complex 2 Protein, Tumor Suppressor Proteins metabolism, Gene Amplification, Medulloblastoma genetics, Mutation, Missense, Proto-Oncogene Proteins c-myc genetics, Signal Transduction genetics, Tumor Suppressor Proteins genetics
- Abstract
Medulloblastoma is the most common malignant brain tumor of childhood. To identify targetable vulnerabilities, we employed inhibitor screening that revealed mTOR inhibitor hypersensitivity in the MYC-overexpressing medulloblastoma cell line, D341. Concomitant exome sequencing unveiled an uncharacterized missense mutation, TSC2
A415V , in these cells. We biochemically demonstrate that the TSC2A415V mutation is functionally deleterious, leading to shortened half-life and proteasome-mediated protein degradation. These data suggest that MYC cooperates with activated kinase pathways, enabling pharmacologic intervention in these treatment refractory tumors. We propose that identification of activated kinase pathways may allow for tailoring targeted therapy to improve survival and treatment-related morbidity in medulloblastoma., (© 2017 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
36. Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic malignancies.
- Author
-
Kurtz SE, Eide CA, Kaempf A, Khanna V, Savage SL, Rofelty A, English I, Ho H, Pandya R, Bolosky WJ, Poon H, Deininger MW, Collins R, Swords RT, Watts J, Pollyea DA, Medeiros BC, Traer E, Tognon CE, Mori M, Druker BJ, and Tyner JW
- Subjects
- Drug Combinations, Hematologic Neoplasms metabolism, Humans, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Leukemia, Myeloid, Acute metabolism, Mutation drug effects, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Antineoplastic Agents therapeutic use, Hematologic Neoplasms drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Leukemia, Myeloid, Acute drug therapy
- Abstract
Translating the genetic and epigenetic heterogeneity underlying human cancers into therapeutic strategies is an ongoing challenge. Large-scale sequencing efforts have uncovered a spectrum of mutations in many hematologic malignancies, including acute myeloid leukemia (AML), suggesting that combinations of agents will be required to treat these diseases effectively. Combinatorial approaches will also be critical for combating the emergence of genetically heterogeneous subclones, rescue signals in the microenvironment, and tumor-intrinsic feedback pathways that all contribute to disease relapse. To identify novel and effective drug combinations, we performed ex vivo sensitivity profiling of 122 primary patient samples from a variety of hematologic malignancies against a panel of 48 drug combinations. The combinations were designed as drug pairs that target nonoverlapping biological pathways and comprise drugs from different classes, preferably with Food and Drug Administration approval. A combination ratio (CR) was derived for each drug pair, and CRs were evaluated with respect to diagnostic categories as well as against genetic, cytogenetic, and cellular phenotypes of specimens from the two largest disease categories: AML and chronic lymphocytic leukemia (CLL). Nearly all tested combinations involving a BCL2 inhibitor showed additional benefit in patients with myeloid malignancies, whereas select combinations involving PI3K, CSF1R, or bromodomain inhibitors showed preferential benefit in lymphoid malignancies. Expanded analyses of patients with AML and CLL revealed specific patterns of ex vivo drug combination efficacy that were associated with select genetic, cytogenetic, and phenotypic disease subsets, warranting further evaluation. These findings highlight the heuristic value of an integrated functional genomic approach to the identification of novel treatment strategies for hematologic malignancies., Competing Interests: Conflict of interest statement: D.A.P. serves on the advisory boards for Pharmacyclics and Gilead. J.W.T. receives research support from Agios, Array Biopharma, Aptose, AstraZeneca, Constellation, Genentech, Gilead, Incyte, Janssen R&D, Seattle Genetics, Syros, and Takeda and is a consultant for Leap Oncology. B.J.D. serves on the advisory boards for Gilead and Roche TCRC. B.J.D. is principal investigator or coinvestigator on Novartis and BMS clinical trials. His institution, Oregon Health & Science University, has contracts with these companies to pay for patient costs, nurse and data manager salaries, and institutional overhead. He does not derive salary, nor does his laboratory receive funds from these contracts. M.W.D. serves on the advisory boards and/or as a consultant for Novartis, Incyte, and BMS and receives research funding from BMS and Gilead. The authors certify that all compounds and combinations tested in this study were chosen without input from any of our industry partners.
- Published
- 2017
- Full Text
- View/download PDF
37. Recurrent cyclin D2 mutations in myeloid neoplasms.
- Author
-
Khanna V, Eide CA, Tognon CE, Maxson JE, Wilmot B, Bottomly D, McWeeney S, Edwards V DK, Druker BJ, and Tyner JW
- Subjects
- Animals, Humans, Mice, NIH 3T3 Cells, Cyclin D2 genetics, Mutation, Myeloproliferative Disorders genetics
- Published
- 2017
- Full Text
- View/download PDF
38. Understanding cancer from the stem cells up.
- Author
-
Eide CA and Druker BJ
- Subjects
- Humans, Stem Cells, Leukemia, Myelogenous, Chronic, BCR-ABL Positive, Neoplasms
- Published
- 2017
- Full Text
- View/download PDF
39. Differentiation status of primary chronic myeloid leukemia cells affects sensitivity to BCR-ABL1 inhibitors.
- Author
-
Pietarinen PO, Eide CA, Ayuda-Durán P, Potdar S, Kuusanmäki H, Andersson EI, Mpindi JP, Pemovska T, Kontro M, Heckman CA, Kallioniemi O, Wennerberg K, Hjorth-Hansen H, Druker BJ, Enserink JM, Tyner JW, Mustjoki S, and Porkka K
- Subjects
- Apoptosis drug effects, Cell Proliferation drug effects, Cohort Studies, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive classification, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Tumor Cells, Cultured, Biomarkers, Tumor metabolism, Cell Differentiation drug effects, Drug Resistance, Neoplasm drug effects, Fusion Proteins, bcr-abl metabolism, High-Throughput Screening Assays methods, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Protein Kinase Inhibitors pharmacology
- Abstract
Tyrosine kinase inhibitors (TKI) are the mainstay treatment of BCR-ABL1-positive leukemia and virtually all patients with chronic myeloid leukemia in chronic phase (CP CML) respond to TKI therapy. However, there is limited information on the cellular mechanisms of response and particularly on the effect of cell differentiation state to TKI sensitivity in vivo and ex vivo/in vitro. We used multiple, independent high-throughput drug sensitivity and resistance testing platforms that collectively evaluated 295 oncology compounds to characterize ex vivo drug response profiles of primary cells freshly collected from newly-diagnosed patients with BCR-ABL1-positive leukemia (n = 40) and healthy controls (n = 12). In contrast to the highly TKI-sensitive cells from blast phase CML and Philadelphia chromosome-positive acute lymphoblastic leukemia, primary CP CML cells were insensitive to TKI therapy ex vivo. Despite maintaining potent BCR-ABL1 inhibitory activity, ex vivo viability of cells was unaffected by TKIs. These findings were validated in two independent patient cohorts and analysis platforms. All CP CML patients under study responded to TKI therapy in vivo. When CP CML cells were sorted based on CD34 expression, the CD34-positive progenitor cells showed good sensitivity to TKIs, whereas the more mature CD34-negative cells were markedly less sensitive. Thus in CP CML, TKIs predominantly target the progenitor cell population while the differentiated leukemic cells (mostly cells from granulocytic series) are insensitive to BCR-ABL1 inhibition. These findings have implications for drug discovery in CP CML and indicate a fundamental biological difference between CP CML and advanced forms of BCR-ABL1-positive leukemia.
- Published
- 2017
- Full Text
- View/download PDF
40. Identification of Interleukin-1 by Functional Screening as a Key Mediator of Cellular Expansion and Disease Progression in Acute Myeloid Leukemia.
- Author
-
Carey A, Edwards DK 5th, Eide CA, Newell L, Traer E, Medeiros BC, Pollyea DA, Deininger MW, Collins RH, Tyner JW, Druker BJ, Bagby GC, McWeeney SK, and Agarwal A
- Subjects
- Bone Marrow drug effects, Bone Marrow metabolism, Cell Proliferation drug effects, Cell Survival drug effects, Humans, Inflammation Mediators metabolism, Intercellular Signaling Peptides and Proteins pharmacology, Interleukin-1beta blood, Interleukin-1beta metabolism, Models, Biological, Monocytes metabolism, Phosphorylation drug effects, Receptors, Interleukin-1 metabolism, Signal Transduction drug effects, Tumor Stem Cell Assay, p38 Mitogen-Activated Protein Kinases metabolism, Disease Progression, Interleukin-1 metabolism, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology
- Abstract
Secreted proteins in the bone marrow microenvironment play critical roles in acute myeloid leukemia (AML). Through an ex vivo functional screen of 94 cytokines, we identified that the pro-inflammatory cytokine interleukin-1 (IL-1) elicited profound expansion of myeloid progenitors in ∼67% of AML patients while suppressing the growth of normal progenitors. Levels of IL-1β and IL-1 receptors were increased in AML patients, and silencing of the IL-1 receptor led to significant suppression of clonogenicity and in vivo disease progression. IL-1 promoted AML cell growth by enhancing p38MAPK phosphorylation and promoting secretion of various other growth factors and inflammatory cytokines. Treatment with p38MAPK inhibitors reversed these effects and recovered normal CD34
+ cells from IL-1-mediated growth suppression. These results highlight the importance of ex vivo functional screening to identify common and actionable extrinsic pathways in genetically heterogeneous malignancies and provide impetus for clinical development of IL-1/IL1R1/p38MAPK pathway-targeted therapies in AML., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
41. Targeting BCL-2 and ABL/LYN in Philadelphia chromosome-positive acute lymphoblastic leukemia.
- Author
-
Leonard JT, Rowley JS, Eide CA, Traer E, Hayes-Lattin B, Loriaux M, Spurgeon SE, Druker BJ, Tyner JW, and Chang BH
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Cell Line, Tumor, Dasatinib administration & dosage, Humans, Imidazoles administration & dosage, Mice, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Pyridazines administration & dosage, STAT5 Transcription Factor metabolism, Translational Research, Biomedical, Xenograft Model Antitumor Assays, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Philadelphia Chromosome, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Protein Kinase Inhibitors administration & dosage, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Sulfonamides administration & dosage, src-Family Kinases antagonists & inhibitors
- Abstract
Treatment of Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph(+)ALL) remains a challenge. Although the addition of targeted tyrosine kinase inhibitors (TKIs) to standard cytotoxic therapy has greatly improved upfront treatment, treatment-related morbidity and mortality remain high. TKI monotherapy provides only temporary responses and renders patients susceptible to the development of TKI resistance. Thus, identifying agents that could enhance the activity of TKIs is urgently needed. Recently, a selective inhibitor of B cell lymphoma 2 (BCL-2), ABT-199 (venetoclax), has shown impressive activity against hematologic malignancies. We demonstrate that the combination of TKIs with venetoclax is highly synergistic in vitro, decreasing cell viability and inducing apoptosis in Ph(+)ALL. Furthermore, the multikinase inhibitors dasatinib and ponatinib appear to have the added advantage of inducing Lck/Yes novel tyrosine kinase (LYN)-mediated proapoptotic BCL-2-like protein 11 (BIM) expression and inhibiting up-regulation of antiapoptotic myeloid cell leukemia 1 (MCL-1), thereby potentially overcoming the development of venetoclax resistance. Evaluation of the dasatinib-venetoclax combination for the treatment of primary Ph(+)ALL patient samples in xenografted immunodeficient mice confirmed the tolerability of this drug combination and demonstrated its superior antileukemic efficacy compared to either agent alone. These data suggest that the combination of dasatinib and venetoclax has the potential to improve the treatment of Ph(+)ALL and should be further evaluated for patient care., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
42. Extreme mutational selectivity of axitinib limits its potential use as a targeted therapeutic for BCR-ABL1-positive leukemia.
- Author
-
Zabriskie MS, Eide CA, Yan D, Vellore NA, Pomicter AD, Savage SL, Druker BJ, Deininger MW, and O'Hare T
- Subjects
- Axitinib, Humans, Fusion Proteins, bcr-abl genetics, Imidazoles therapeutic use, Indazoles therapeutic use, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Mutation genetics, Protein Kinase Inhibitors therapeutic use
- Published
- 2016
- Full Text
- View/download PDF
43. A Novel Crizotinib-Resistant Solvent-Front Mutation Responsive to Cabozantinib Therapy in a Patient with ROS1-Rearranged Lung Cancer.
- Author
-
Drilon A, Somwar R, Wagner JP, Vellore NA, Eide CA, Zabriskie MS, Arcila ME, Hechtman JF, Wang L, Smith RS, Kris MG, Riely GJ, Druker BJ, O'Hare T, Ladanyi M, and Davare MA
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma of Lung, Antigens, Differentiation, B-Lymphocyte genetics, Cell Line, Tumor, Clinical Trials, Phase II as Topic, Crizotinib, Drug Resistance, Neoplasm genetics, Female, Histocompatibility Antigens Class II genetics, Humans, Middle Aged, Mutation genetics, Oncogenes drug effects, Oncogenes genetics, Anilides therapeutic use, Drug Resistance, Neoplasm drug effects, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Mutation drug effects, Protein-Tyrosine Kinases genetics, Proto-Oncogene Proteins genetics, Pyrazoles therapeutic use, Pyridines therapeutic use
- Abstract
Purpose: Rearranged ROS1 is a crizotinib-sensitive oncogenic driver in lung cancer. The development of acquired resistance, however, poses a serious clinical challenge. Consequently, experimental and clinical validation of resistance mechanisms and potential second-line therapies is essential., Experimental Design: We report the discovery of a novel, solvent-front ROS1(D2033N) mutation in a patient with CD74-ROS1-rearranged lung adenocarcinoma and acquired resistance to crizotinib. Crizotinib resistance of CD74-ROS1(D2033N) was functionally evaluated using cell-based assays and structural modeling., Results: In biochemical and cell-based assays, the CD74-ROS1(D2033N) mutant demonstrated significantly decreased sensitivity to crizotinib. Molecular dynamics simulation revealed compromised crizotinib binding due to drastic changes in the electrostatic interaction between the D2033 residue and crizotinib and reorientation of neighboring residues. In contrast, cabozantinib binding was unaffected by the D2033N substitution, and inhibitory potency against the mutant was retained. Notably, cabozantinib treatment resulted in a rapid clinical and near-complete radiographic response in this patient., Conclusions: These results provide the first example of successful therapeutic intervention with targeted therapy to overcome crizotinib resistance in a ROS1-rearranged cancer. Clin Cancer Res; 22(10); 2351-8. ©2015 AACR., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
44. IRS2 silencing increases apoptosis and potentiates the effects of ruxolitinib in JAK2V617F-positive myeloproliferative neoplasms.
- Author
-
de Melo Campos P, Machado-Neto JA, Eide CA, Savage SL, Scopim-Ribeiro R, da Silva Souza Duarte A, Favaro P, Lorand-Metze I, Costa FF, Tognon CE, Druker BJ, Olalla Saad ST, and Traina F
- Subjects
- Adult, Aged, Aged, 80 and over, Blotting, Western, Case-Control Studies, Cell Proliferation drug effects, Female, Fluorescent Antibody Technique, Follow-Up Studies, Gene Silencing, Humans, Immunoenzyme Techniques, Immunoprecipitation, Insulin Receptor Substrate Proteins genetics, Insulin Receptor Substrate Proteins metabolism, Janus Kinase 2 metabolism, Male, Middle Aged, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Neoplasm Staging, Nitriles, Prognosis, Pyrimidines, RNA, Messenger genetics, RNA, Small Interfering genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Young Adult, Apoptosis drug effects, Insulin Receptor Substrate Proteins antagonists & inhibitors, Janus Kinase 2 genetics, Mutation genetics, Myeloproliferative Disorders pathology, Pyrazoles pharmacology
- Abstract
The recurrent V617F mutation in JAK2 (JAK2V617F) has emerged as the primary contributor to the pathogenesis of myeloproliferative neoplasms (MPN). However, the lack of complete response in most patients treated with the JAK1/2 inhibitor, ruxolitinib, indicates the need for identifying pathways that cooperate with JAK2. Activated JAK2 was found to be associated with the insulin receptor substrate 2 (IRS2) in non-hematological cells. We identified JAK2/IRS2 binding in JAK2V617F HEL cells, but not in the JAK2WT U937 cell line. In HEL cells, IRS2 silencing decreased STAT5 phosphorylation, reduced cell viability and increased apoptosis; these effects were enhanced when IRS2 silencing was combined with ruxolitinib. In U937 cells, IRS2 silencing neither reduced cell viability nor induced apoptosis. IRS1/2 pharmacological inhibition in primary MPN samples reduced cell viability in JAK2V617F-positive but not JAK2WT specimens; combination with ruxolitinib had additive effects. IRS2 expression was significantly higher in CD34+ cells from essential thrombocythemia patients compared to healthy donors, and in JAK2V617F MPN patients when compared to JAK2WT. Our data indicate that IRS2 is a binding partner of JAK2V617F in MPN. IRS2 contributes to increased cell viability and reduced apoptosis in JAK2-mutated cells. Combined pharmacological inhibition of IRS2 and JAK2 may have a potential clinical application in MPN.
- Published
- 2016
- Full Text
- View/download PDF
45. The Colony-Stimulating Factor 3 Receptor T640N Mutation Is Oncogenic, Sensitive to JAK Inhibition, and Mimics T618I.
- Author
-
Maxson JE, Luty SB, MacManiman JD, Paik JC, Gotlib J, Greenberg P, Bahamadi S, Savage SL, Abel ML, Eide CA, Loriaux MM, Stevens EA, and Tyner JW
- Subjects
- Amino Acid Substitution, Animals, Bone Marrow pathology, Bone Marrow Transplantation, Cell Line, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, DNA Mutational Analysis, Disease Models, Animal, Female, Glycosylation, Humans, Leukemia drug therapy, Leukemia genetics, Leukemia metabolism, Leukemia mortality, Leukemia pathology, Ligands, Male, Mice, Middle Aged, Receptors, Colony-Stimulating Factor metabolism, Codon, Janus Kinases antagonists & inhibitors, Mutation, Protein Kinase Inhibitors pharmacology, Receptors, Colony-Stimulating Factor genetics
- Abstract
Purpose: Colony-stimulating factor 3 receptor (CSF3R) mutations have been identified in the majority of chronic neutrophilic leukemia (CNL) and a smaller percentage of atypical chronic myeloid leukemia (aCML) cases. Although CSF3R point mutations (e.g., T618I) are emerging as key players in CNL/aCML, the significance of rarer CSF3R mutations is unknown. In this study, we assess the importance of the CSF3R T640N mutation as a marker of CNL/aCML and potential therapeutic target., Experimental Design: Sanger sequencing of leukemia samples was performed to identify CSF3R mutations in CNL and aCML. The oncogenicity of the CSF3R T640N mutation relative to the T618I mutation was assessed by cytokine independent growth assays and by mouse bone marrow transplant. Receptor dimerization and O-glycosylation of the mutants was assessed by Western blot, and JAK inhibitor sensitivity was assessed by colony assay., Results: Here, we identify a CSF3R T640N mutation in two patients with CNL/aCML, one of whom was originally diagnosed with MDS and acquired the T640N mutation upon evolution of disease to aCML. The T640N mutation is oncogenic in cellular transformation assays and an in vivo mouse bone marrow transplantation model. It exhibits many similar phenotypic features to T618I, including ligand independence and altered patterns of O-glycosylation--despite the transmembrane location of T640 preventing access by GalNAc transferase enzymes. Cells transformed by the T640N mutation are sensitive to JAK kinase inhibition to a similar degree as cells transformed by CSF3R T618I., Conclusions: Because of its similarities to CSF3R T618I, the T640N mutation likely has diagnostic and therapeutic relevance in CNL/aCML., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
46. Src and STAT3 inhibitors synergize to promote tumor inhibition in renal cell carcinoma.
- Author
-
Lue HW, Cole B, Rao SA, Podolak J, Van Gaest A, King C, Eide CA, Wilmot B, Xue C, Spellman PT, Heiser LM, Tyner JW, and Thomas GV
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Animals, Apoptosis drug effects, Carcinoma, Renal Cell enzymology, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Drug Synergism, Gene Expression Regulation, Neoplastic, Kidney Neoplasms enzymology, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Mice, Molecular Targeted Therapy, Phosphoproteins genetics, Phosphoproteins metabolism, STAT3 Transcription Factor metabolism, Signal Transduction drug effects, Time Factors, Transcription Factors, Transcription, Genetic, Tumor Burden drug effects, Xenograft Model Antitumor Assays, YAP-Signaling Proteins, src-Family Kinases metabolism, Antineoplastic Combined Chemotherapy Protocols pharmacology, Benzamides pharmacology, Carcinoma, Renal Cell drug therapy, Dasatinib pharmacology, Kidney Neoplasms drug therapy, Protein Kinase Inhibitors pharmacology, Pyrimidines pharmacology, STAT3 Transcription Factor antagonists & inhibitors, src-Family Kinases antagonists & inhibitors
- Abstract
The intracytoplasmic tyrosine kinase Src serves both as a conduit and a regulator for multiple processes required for the proliferation and survival cancer cells. In some cancers, Src engages with receptor tyrosine kinases to mediate downstream signaling and in other cancers, it regulates gene expression. Src therefore represents a viable oncologic target. However, clinical responses to Src inhibitors, such as dasatinib have been disappointing to date. We identified Stat3 signaling as a potential bypass mechanism that enables renal cell carcinoma (RCC) cells to escape dasatinib treatment. Combined Src-Stat3 inhibition using dasatinib and CYT387 (a JAK/STAT inhibitor) synergistically reduced cell proliferation and increased apoptosis in RCC cells. Moreover, dasatinib and CYT387 combine to suppress YAP1, a transcriptional co-activator that promotes cell proliferation, survival and organ size. Importantly, this combination was well tolerated, and caused marked tumor inhibition in RCC xenografts. These results suggest that combination therapy with inhibitors of Stat3 signaling may be a useful therapeutic approach to increase the efficacy of Src inhibitors.
- Published
- 2015
- Full Text
- View/download PDF
47. Structural insight into selectivity and resistance profiles of ROS1 tyrosine kinase inhibitors.
- Author
-
Davare MA, Vellore NA, Wagner JP, Eide CA, Goodman JR, Drilon A, Deininger MW, O'Hare T, and Druker BJ
- Subjects
- Anaplastic Lymphoma Kinase, Crizotinib, Drug Discovery methods, Humans, Immunoblotting, In Vitro Techniques, Molecular Dynamics Simulation, Protein Binding, Protein Conformation, Protein-Tyrosine Kinases chemistry, Proto-Oncogene Proteins chemistry, Pyrazoles, Quinolines, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Receptor Protein-Tyrosine Kinases chemistry, Anilides pharmacology, Antineoplastic Agents pharmacology, Carcinoma, Non-Small-Cell Lung drug therapy, Drug Resistance, Neoplasm physiology, Models, Molecular, Protein-Tyrosine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors, Pyridines pharmacology
- Abstract
Oncogenic ROS1 fusion proteins are molecular drivers in multiple malignancies, including a subset of non-small cell lung cancer (NSCLC). The phylogenetic proximity of the ROS1 and anaplastic lymphoma kinase (ALK) catalytic domains led to the clinical repurposing of the Food and Drug Administration (FDA)-approved ALK inhibitor crizotinib as a ROS1 inhibitor. Despite the antitumor activity of crizotinib observed in both ROS1- and ALK-rearranged NSCLC patients, resistance due to acquisition of ROS1 or ALK kinase domain mutations has been observed clinically, spurring the development of second-generation inhibitors. Here, we profile the sensitivity and selectivity of seven ROS1 and/or ALK inhibitors at various levels of clinical development. In contrast to crizotinib's dual ROS1/ALK activity, cabozantinib (XL-184) and its structural analog foretinib (XL-880) demonstrate a striking selectivity for ROS1 over ALK. Molecular dynamics simulation studies reveal structural features that distinguish the ROS1 and ALK kinase domains and contribute to differences in binding site and kinase selectivity of the inhibitors tested. Cell-based resistance profiling studies demonstrate that the ROS1-selective inhibitors retain efficacy against the recently reported CD74-ROS1(G2032R) mutant whereas the dual ROS1/ALK inhibitors are ineffective. Taken together, inhibitor profiling and stringent characterization of the structure-function differences between the ROS1 and ALK kinase domains will facilitate future rational drug design for ROS1- and ALK-driven NSCLC and other malignancies.
- Published
- 2015
- Full Text
- View/download PDF
48. Functional RNAi screen targeting cytokine and growth factor receptors reveals oncorequisite role for interleukin-2 gamma receptor in JAK3-mutation-positive leukemia.
- Author
-
Agarwal A, MacKenzie RJ, Eide CA, Davare MA, Watanabe-Smith K, Tognon CE, Mongoue-Tchokote S, Park B, Braziel RM, Tyner JW, and Druker BJ
- Subjects
- Animals, Binding Sites, Cell Line, Tumor, Humans, Interleukin Receptor Common gamma Subunit antagonists & inhibitors, Interleukin Receptor Common gamma Subunit genetics, Janus Kinase 3 metabolism, Leukemia metabolism, Leukemia pathology, Mice, Molecular Sequence Data, Mutation, Phosphorylation, Signal Transduction, Cell Transformation, Neoplastic genetics, Interleukin Receptor Common gamma Subunit metabolism, Janus Kinase 3 genetics, Leukemia genetics, RNA, Small Interfering pharmacology
- Abstract
To understand the role of cytokine and growth factor receptor-mediated signaling in leukemia pathogenesis, we designed a functional RNA interference (RNAi) screen targeting 188 cytokine and growth factor receptors that we found highly expressed in primary leukemia specimens. Using this screen, we identified interleukin-2 gamma receptor (IL2Rγ) as a critical growth determinant for a JAK3(A572V) mutation-positive acute myeloid leukemia cell line. We observed that knockdown of IL2Rγ abrogates phosphorylation of JAK3 and downstream signaling molecules, JAK1, STAT5, MAPK and pS6 ribosomal protein. Overexpression of IL2Rγ in murine cells increased the transforming potential of activating JAK3 mutations, whereas absence of IL2Rγ completely abrogated the clonogenic potential of JAK3(A572V), as well as the transforming potential of additional JAK3-activating mutations such as JAK3(M511I). In addition, mutation at the IL2Rγ interaction site in the FERM domain of JAK3 (Y100C) completely abrogated JAK3-mediated leukemic transformation. Mechanistically, we found IL2Rγ contributes to constitutive JAK3 mutant signaling by increasing JAK3 expression and phosphorylation. Conversely, we found that mutant, but not wild-type JAK3, increased the expression of IL2Rγ, indicating IL2Rγ and JAK3 contribute to constitutive JAK/STAT signaling through their reciprocal regulation. Overall, we demonstrate a novel role for IL2Rγ in potentiating oncogenesis in the setting of JAK3-mutation-positive leukemia. In addition, our study highlights an RNAi-based functional assay that can be used to facilitate the identification of non-kinase cytokine and growth factor receptor targets for inhibiting leukemic cell growth.
- Published
- 2015
- Full Text
- View/download PDF
49. Chronic myeloid leukemia: advances in understanding disease biology and mechanisms of resistance to tyrosine kinase inhibitors.
- Author
-
Eide CA and O'Hare T
- Subjects
- Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl metabolism, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive enzymology, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Molecular Targeted Therapy, Mutation, Protein Kinase Inhibitors chemistry, Protein-Tyrosine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases metabolism, Antineoplastic Agents therapeutic use, Drug Resistance, Neoplasm drug effects, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Protein Kinase Inhibitors therapeutic use
- Abstract
The successful implementation of tyrosine kinase inhibitors (TKIs) for the treatment of chronic myeloid leukemia (CML) remains a flagship for molecularly targeted therapy in cancer. This focused review highlights critical elements of the underlying biology of CML and provides a summary of the molecular mechanisms that lead to TKI resistance: BCR-ABL1 mutation-based resistance and therapy escape through alternative pathway activation despite inhibition of BCR-ABL1 tyrosine kinase activity. We direct attention to the most current manifestations of these issues, including emergence of pan-TKI-resistant BCR-ABL1 compound mutants, new strategies for identification and therapeutic targeting of alternative pathways, and the exciting, controversial topic of cessation of TKI therapy leading to durable treatment-free remissions for a subset of patients. Further gains in our understanding of the biology of Philadelphia chromosome-positive (Ph-positive) leukemia and mechanisms of resistance to BCR-ABL1 TKIs will benefit patients and also provide a blueprint for similar discovery in other cancers.
- Published
- 2015
- Full Text
- View/download PDF
50. Therapeutically Targetable ALK Mutations in Leukemia.
- Author
-
Maxson JE, Davare MA, Luty SB, Eide CA, Chang BH, Loriaux MM, Tognon CE, Bottomly D, Wilmot B, McWeeney SK, Druker BJ, and Tyner JW
- Subjects
- Anaplastic Lymphoma Kinase, Animals, Cell Line, Tumor, Crizotinib, Drug Resistance, Neoplasm genetics, High-Throughput Nucleotide Sequencing, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology, Mice, Mutation, Precision Medicine, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Protein Kinase Inhibitors administration & dosage, Pyrazoles administration & dosage, Pyridines administration & dosage, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Leukemia, Myeloid, Acute genetics, Molecular Targeted Therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Receptor Protein-Tyrosine Kinases genetics
- Abstract
Genome sequencing is revealing a vast mutational landscape in leukemia, offering new opportunities for treatment with targeted therapy. Here, we identify two patients with acute myelogenous leukemia and B-cell acute lymphoblastic leukemia whose tumors harbor point mutations in the ALK kinase. The mutations reside in the extracellular domain of ALK and are potently transforming in cytokine-independent cellular assays and primary mouse bone marrow colony formation studies. Strikingly, both mutations conferred sensitivity to ALK kinase inhibitors, including the FDA-approved drug crizotinib. On the basis of our results, we propose that tumors harboring ALK mutations may be therapeutically tractable for personalized treatment of certain aggressive leukemias with ALK inhibitors., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.