82 results on '"Kathy L. McGraw"'
Search Results
2. Somatic gene mutations expose cytoplasmic DNA to co-opt the cGAS/STING/NLRP3 axis in myelodysplastic syndromes
- Author
-
Amy F. McLemore, Hsin-An Hou, Benjamin S. Meyer, Nghi B. Lam, Grace A. Ward, Amy L. Aldrich, Matthew A. Rodrigues, Alexis Vedder, Ling Zhang, Eric Padron, Nicole D. Vincelette, David A. Sallman, Omar Abdel-Wahab, Alan F. List, and Kathy L. McGraw
- Subjects
Hematology ,Oncology ,Medicine - Abstract
NLRP3 inflammasome and IFN-stimulated gene (ISG) induction are key biological drivers of ineffective hematopoiesis and inflammation in myelodysplastic syndromes (MDSs). Gene mutations involving mRNA splicing and epigenetic regulatory pathways induce inflammasome activation and myeloid lineage skewing in MDSs through undefined mechanisms. Using immortalized murine hematopoietic stem and progenitor cells harboring these somatic gene mutations and primary MDS BM specimens, we showed accumulation of unresolved R-loops and micronuclei with concurrent activation of the cytosolic sensor cyclic GMP-AMP synthase. Cyclic GMP-AMP synthase/stimulator of IFN genes (cGAS/STING) signaling caused ISG induction, NLRP3 inflammasome activation, and maturation of the effector protease caspase-1. Deregulation of RNA polymerase III drove cytosolic R-loop generation, which upon inhibition, extinguished ISG and inflammasome response. Mechanistically, caspase-1 degraded the master erythroid transcription factor, GATA binding protein 1, provoking anemia and myeloid lineage bias that was reversed by cGAS inhibition in vitro and in Tet2–/– hematopoietic stem and progenitor cell–transplanted mice. Together, these data identified a mechanism by which functionally distinct mutations converged upon the cGAS/STING/NLRP3 axis in MDS, directing ISG induction, pyroptosis, and myeloid lineage skewing.
- Published
- 2022
- Full Text
- View/download PDF
3. Non-del(5q) myelodysplastic syndromes–associated loci detected by SNP-array genome-wide association meta-analysis
- Author
-
Kathy L. McGraw, Chia-Ho Cheng, Y. Ann Chen, Hsin-An Hou, Björn Nilsson, Giulio Genovese, Thomas Cluzeau, Andrea Pellagatti, Bartlomiej P. Przychodzen, Mar Mallo, Leonor Arenillas, Azim Mohamedali, Lionel Adès, David A. Sallman, Eric Padron, Lubomir Sokol, Chimene Moreilhon, Sophie Raynaud, Hwei-Fang Tien, Jacqueline Boultwood, Benjamin L. Ebert, Francesc Sole, Pierre Fenaux, Ghulam J. Mufti, Jaroslaw P. Maciejewski, Peter A. Kanetsky, and Alan F. List
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Myelodysplastic syndromes (MDS) are hematopoietic stem cell malignancies. Known predisposing factors to adult MDS include rare germline mutations, cytotoxic therapy, age-related clonal hematopoiesis, and autoimmune or chronic inflammatory disorders. To date, no published studies characterizing MDS-associated germline susceptibility polymorphisms exist. We performed a genome-wide association study of 2 sample sets (555 MDS cases vs 2964 control subjects; 352 MDS cases vs 2640 control subjects) in non-del(5q) MDS cases of European genomic ancestry. Meta-analysis identified 8 MDS-associated loci at 1q31.1 (PLA2G4A), 3p14.1 (FAM19A4), 5q21.3 (EFNA5), 6p21.33, 10q23.1 (GRID1), 12q24.32, 15q26.1, and 20q13.12 (EYA2) that approached genome-wide significance. Gene expression for 5 loci that mapped within or near genes was significantly upregulated in MDS bone marrow cells compared with those of control subjects (P < .01). Higher PLA2G4A expression and lower EYA2 expression were associated with poorer overall survival (P = .039 and P = .037, respectively). Higher PLA2G4A expression is associated with mutations in NRAS (P < .001), RUNX1 (P = .012), ASXL1 (P = .007), and EZH2 (P = .038), all of which are known to contribute to MDS development. EYA2 expression was an independently favorable risk factor irrespective of age, sex, and Revised International Scoring System score (relative risk, 0.67; P = .048). Notably, these genes have regulatory roles in innate immunity, a critical driver of MDS pathogenesis. EYA2 overexpression induced innate immune activation, whereas EYA2 inhibition restored colony-forming potential in primary MDS cells indicative of hematopoietic restoration and possible clinical relevance. In conclusion, among 8 suggestive MDS-associated loci, 5 map to genes upregulated in MDS with functional roles in innate immunity and potential biological relevance to MDS.
- Published
- 2019
- Full Text
- View/download PDF
4. Dual pyroptotic biomarkers predict erythroid response in lower-risk non-del(5q) myelodysplastic syndromes treated with lenalidomide and recombinant erythropoietin
- Author
-
Chen Wang, Kathy L. McGraw, Amy F. McLemore, Rami Komrokji, Ashley A. Basiorka, Najla Al Ali, Jeffrey E. Lancet, Eric Padron, Olivier Kosmider, Michaela Fontenay, Pierre Fenaux, Alan F. List, and David A. Sallman
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2021
- Full Text
- View/download PDF
5. Pro-inflammatory proteins S100A9 and tumor necrosis factor-α suppress erythropoietin elaboration in myelodysplastic syndromes
- Author
-
Thomas Cluzeau, Kathy L. McGraw, Brittany Irvine, Erico Masala, Lionel Ades, Ashley A. Basiorka, Jaroslaw Maciejewski, Patrick Auberger, Sheng Wei, Pierre Fenaux, Valeria Santini, and Alan List
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Accumulating evidence implicates innate immune activation in the pathobiology of myelodysplastic syndromes. A key myeloid-related inflammatory protein, S100A9, serves as a Toll-like receptor ligand regulating tumor necrosis factor-α and interleukin-1β production. The role of myelodysplastic syndrome-related inflammatory proteins in endogenous erythropoietin regulation and response to erythroid-stimulating agents or lenalidomide has not been investigated. The HepG2 hepatoma cell line was used to investigate in vitro erythropoietin elaboration. Serum samples collected from 311 patients with myelodysplastic syndrome were investigated (125 prior to treatment with erythroid-stimulating agents and 186 prior to lenalidomide therapy). Serum concentrations of S100A9, S100A8, tumor necrosis factor-α, interleukin-1β and erythropoietin were analyzed by enzyme-linked immunosorbent assay. Using erythropoietin-producing HepG2 cells, we show that S100A9, tumor necrosis factor-α and interleukin-1β suppress transcription and cellular elaboration of erythropoietin. Pre-incubation with lenalidomide significantly diminished suppression of erythropoietin production by S100A9 or tumor necrosis factor-α. Moreover, in peripheral blood mononuclear cells from patients with myelodysplastic syndromes, lenalidomide significantly reduced steady-state S100A9 generation (P=0.01) and lipopolysaccharide-induced tumor necrosis factor-α elaboration (P=0.002). Enzyme-linked immunosorbent assays of serum from 316 patients with non-del(5q) myelodysplastic syndromes demonstrated a significant inverse correlation between tumor necrosis factor-α and erythropoietin concentrations (P=0.006), and between S100A9 and erythropoietin (P=0.01). Moreover, baseline serum tumor necrosis factor-α concentration was significantly higher in responders to erythroid-stimulating agents (P=0.03), whereas lenalidomide responders had significantly lower tumor necrosis factor-α and higher S100A9 serum concentrations (P=0.03). These findings suggest that S100A9 and its nuclear factor-κB transcriptional target, tumor necrosis factor-α, directly suppress erythropoietin elaboration in myelodysplastic syndromes. These cytokines may serve as rational biomarkers of response to lenalidomide and erythroid-stimulating agent treatments. Therapeutic strategies that either neutralize or suppress S100A9 may improve erythropoiesis in patients with myelodysplastic syndromes.
- Published
- 2017
- Full Text
- View/download PDF
6. Immunohistochemical pattern of p53 is a measure of TP53 mutation burden and adverse clinical outcome in myelodysplastic syndromes and secondary acute myeloid leukemia
- Author
-
Kathy L. McGraw, Johnny Nguyen, Rami S. Komrokji, David Sallman, Najla H. Al Ali, Eric Padron, Jeffrey E. Lancet, Lynn C. Moscinski, Alan F. List, and Ling Zhang
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2016
- Full Text
- View/download PDF
7. Supplementary Data from Loss of Function of DOCK4 in Myelodysplastic Syndromes Stem Cells is Restored by Inhibitors of DOCK4 Signaling Networks
- Author
-
Amittha Wickrema, Amit Verma, Alan F. List, Sandeep Gurbuxani, Kathy L. McGraw, Tushar D. Bhagat, Hui Liu, Shanisha Gordon-Mitchell, Gaurav S. Choudhary, Jong Jin Jeong, Wen-Liang Kuo, and Sriram Sundaravel
- Abstract
Supplemental figure 2: A) Flow cytometry analysis showing that the TF1 erythroleukemia cells express high levels of CD34 marker on their cell surface. B) Twenty four hours post DOCK4 siRNA nucleofection, cells were used for immunoblot analysis for DOCK4 expression. Data are mean {plus minus}SEM from five independent experiments. C) Twenty four hours post DOCK4 knockdown, TF1 cells were briefly deprived of cytokines and then re-exposed to a cytokine cocktail (Stem cell factor (SCF), Interleukin-3 (IL-3) and Granulocyte-Macrophage colony stimulating factor (GM-CSF)) for 15 mins. Samples were resolved by immunoblotting for phospho-LYN (Y397), phospho-SHIP1 (Y1021) and phospho-SHP1 (Y536). Same membranes were probed with total LYN, SHIP1 and SHP1 antibodies as loading controls. D, E and F) Quantitation of the levels of phospho-LYN (Y397) (N=3), phospho-SHIP1 (Y1021) (N=5) and phospho-SHP1 (Y536) (N=4) respectively in TF1 cells from control and DOCK4 knocked down samples (*P < 0.05; Student's t test). Data are mean {plus minus}SEM from independent experiments as specified. G) Twenty four hours post DOCK4 knockdown HSCs were briefly deprived and exposed to 5 cytokine cocktails (TPO, SCF, FLT3 ligand, IL3 and IL6) for 15 mins. Following cytokine exposure, LYN kinase activity assay was performed to examine the endogeneous LYN kinase activity in HSCs expressing normal and reduced levels of DOCK4 (*P < 0.05; Student's t test). Data are represented as mean {plus minus}SEM from three technical replicates. H) Quantitation of the levels of phospho-SHIP1 (Y1021) in the in vitro kinase reaction products (***P < 0.0005; One way ANOVA). Data are mean {plus minus}SEM from three independent experiments. I) Quantitation of the levels of phospho-SHP1 (Y536) in the in vitro kinase reaction products (***P < 0.0005; One way ANOVA). Data are mean {plus minus}SEM from three independent experiments. J, K and L) Quantitation of the levels of phospho-LYN (Y397), phospho-SHIP1 (Y1021), and phospho-SHP1 (Y536) respectively in primary human HSCs treated with increasing doses of LYN/Src inhibitor, RK20449. Data are mean {plus minus}SEM from three biological replicates (****P < 0.00005, One way ANOVA; ns-not significant). M) LYN kinase activity assay was performed using recombinant LYN kinase and increasing amounts of recombinant DOCK4 C-terminus. JAK2 kinase and LYN/Src inhibitor were used as controls. Data are mean {plus minus}SEM from three independent experiments. (**P < 0.005; One way ANOVA). N) An in vitro binding assay was performed using flag-tagged DOCK4 C-terminus (D4CT) and recombinant LYN kinase to determine whether LYN kinase interacted with DOCK4-C-terminus. Samples were immunoprecipitated with anti-Flag antibody followed by immunoblot analysis using anti-Flag and anti-LYN antibodies.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary methods from Loss of Function of DOCK4 in Myelodysplastic Syndromes Stem Cells is Restored by Inhibitors of DOCK4 Signaling Networks
- Author
-
Amittha Wickrema, Amit Verma, Alan F. List, Sandeep Gurbuxani, Kathy L. McGraw, Tushar D. Bhagat, Hui Liu, Shanisha Gordon-Mitchell, Gaurav S. Choudhary, Jong Jin Jeong, Wen-Liang Kuo, and Sriram Sundaravel
- Abstract
Supplementary methods
- Published
- 2023
- Full Text
- View/download PDF
9. Data from Loss of Function of DOCK4 in Myelodysplastic Syndromes Stem Cells is Restored by Inhibitors of DOCK4 Signaling Networks
- Author
-
Amittha Wickrema, Amit Verma, Alan F. List, Sandeep Gurbuxani, Kathy L. McGraw, Tushar D. Bhagat, Hui Liu, Shanisha Gordon-Mitchell, Gaurav S. Choudhary, Jong Jin Jeong, Wen-Liang Kuo, and Sriram Sundaravel
- Abstract
Purpose:Myelodysplastic syndromes (MDS) with deletion of chromosome 7q/7 [-7/(del)7q MDS] is associated with worse outcomes and needs novel insights into pathogenesis. Reduced expression of signaling protein dedicator of cytokinesis 4 (DOCK4) in patients with -7/(del)7q MDS leads to a block in hematopoietic stem cell (HSC) differentiation. Identification of targetable signaling networks downstream of DOCK4 will provide means to restore hematopoietic differentiation in MDS.Experimental Design: We utilized phosphoproteomics approaches to identify signaling proteins perturbed as a result of reduced expression of DOCK4 in human HSCs and tested their functional significance in primary model systems.Results:We demonstrate that reduced levels of DOCK4 lead to increased global tyrosine phosphorylation of proteins in primary human HSCs. LYN kinase and phosphatases INPP5D (SHIP1) and PTPN6 (SHP1) displayed greatest levels of tyrosine phosphorylation when DOCK4 expression levels were reduced using DOCK4-specific siRNA. Our data also found that increased phosphorylation of SHIP1 and SHP1 phosphatases were due to LYN kinase targeting these phosphatases as substrates. Increased migration and impediment of HSC differentiation were consequences of these signaling alterations. Pharmacologic inhibition of SHP1 reversed these functional aberrations in HSCs expressing low DOCK4 levels. In addition, differentiation block seen in DOCK4 haplo-insufficient [-7/(del)7q] MDS was rescued by inhibition of SHP1 phosphatase.Conclusions:LYN kinase and phosphatases SHP1 and SHIP1 are perturbed when DOCK4 expression levels are low. Inhibition of SHP1 promotes erythroid differentiation in healthy HSCs and in -7/(del)7q MDS samples with low DOCK4 expression. Inhibitors of LYN, SHP1 and SHIP1 also abrogated increased migratory properties in HSCs expressing reduced levels of DOCK4.
- Published
- 2023
- Full Text
- View/download PDF
10. Arginine metabolism regulates human erythroid differentiation through hypusination of eIF5A
- Author
-
Pedro Gonzalez-Menendez, Ira Phadke, Meagan E Olive, Axel Joly, Julien Papoin, Hongxia Yan, Jérémy Galtier, Jessica Platon, Sun Woo Sophie Kang, Kathy L. McGraw, Marie Daumur, Marie Pouzolles, Taisuke Kondo, Stéphanie Boireau, Franciane Paul, David J Young, Sylvain Lamure, Raghavendra G Mirmira, Anu Narla, Guillaume Cartron, Cynthia E. Dunbar, Myriam Boyer-Clavel, Natalie Porat-Shliom, Valerie Dardalhon, Valerie S Zimmermann, Marc Sitbon, Thomas Dever, Narla Mohandas, Lydie M Da Costa, Namrata D. Udeshi, Lionel Blanc, Sandrina Kinet, and Naomi Taylor
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Metabolic programs contribute to hematopoietic stem and progenitor cell (HSPC) fate, but it is not known whether the metabolic regulation of protein synthesis controls HSPC differentiation. Here, we show that SLC7A1/CAT1-dependent arginine uptake and its catabolism to the polyamine spermidine control human erythroid specification of HSPCs via activation of the eukaryotic translation initiation factor 5A (eIF5A). eIF5A activity is dependent on its hypusination, a post-translational modification resulting from the conjugation of the aminobutyl moiety of spermidine to lysine. Notably, attenuation of hypusine synthesis in erythroid progenitors--by inhibition of deoxyhypusine synthase--abrogates erythropoiesis but not myeloid cell differentiation. Proteomic profiling reveals mitochondrial translation to be a critical target of hypusinated eIF5A and accordingly, progenitors with decreased hypusine activity exhibit diminished oxidative phosphorylation. This impacted pathway is critical for eIF5A-regulated erythropoiesis as interventions augmenting mitochondrial function partially rescue human erythropoiesis under conditions of attenuated hypusination. Levels of mitochondrial ribosomal proteins were especially sensitive to the loss of hypusine and we find that the ineffective erythropoiesis linked to haploinsufficiency of RPS14 in del(5q) myelodysplastic syndrome is associated with a diminished pool of hypusinated eIF5A. Moreover, patients with RPL11-haploinsufficient Diamond-Blackfan anemia as well as CD34+ progenitors with downregulated RPL11 exhibit a markedly decreased hypusination in erythroid progenitors, concomitant with a loss of mitochondrial metabolism. Thus, eIF5A-dependent protein synthesis regulates human erythropoiesis and our data reveal a novel role for RPs in controlling eIF5A hypusination in HSPC, synchronizing mitochondrial metabolism with erythroid differentiation.
- Published
- 2023
- Full Text
- View/download PDF
11. Pyroptosis Markers in Human Primary Specimens: Quantification of Intracellular ASC Specks by Imaging Flow Cytometry and Extracellular Oxidized Mitochondrial by ELISA
- Author
-
Amy F. McLemore, Silvia L. Lage, Venina M. Dominical, Matthew A. Rodrigues, and Kathy L. McGraw
- Published
- 2023
- Full Text
- View/download PDF
12. Eprenetapopt (APR-246) and Azacitidine in TP53-Mutant Myelodysplastic Syndromes
- Author
-
Guillermo Garcia-Manero, Greg Korbel, Gail J. Roboz, Jiqiang Yao, David P. Steensma, Amy F McLemore, Pierre Fenaux, Kathy L. McGraw, David A. Sallman, Alan F. List, Eyal C. Attar, Jeffrey E. Lancet, Hagop M. Kantarjian, Mikkael A. Sekeres, John Puskas, Lisa A Nardelli, Rami S. Komrokji, Eric Padron, Qianxing Mo, Kendra Sweet, Amy E. DeZern, Ling Zhang, Najla Al Ali, and Thomas Cluzeau
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Extramural ,Myelodysplastic syndromes ,Mutant ,Azacitidine ,Complete remission ,medicine.disease ,Clinical trial ,Multicenter study ,hemic and lymphatic diseases ,Internal medicine ,Mutation (genetic algorithm) ,medicine ,business ,medicine.drug - Abstract
PURPOSE Approximately 20% of patients with TP53-mutant myelodysplastic syndromes (MDS) achieve complete remission (CR) with hypomethylating agents. Eprenetapopt (APR-246) is a novel, first-in-class, small molecule that restores wild-type p53 functions in TP53-mutant cells. METHODS This was a phase Ib/II study to determine the safety, recommended phase II dose, and efficacy of eprenetapopt administered in combination with azacitidine in patients with TP53-mutant MDS or acute myeloid leukemia (AML) with 20%-30% marrow blasts (ClinicalTrials.gov identifier: NCT03072043 ). RESULTS Fifty-five patients (40 MDS, 11 AML, and four MDS/myeloproliferative neoplasms) with at least one TP53 mutation were treated. The overall response rate was 71% with 44% achieving CR. Of patients with MDS, 73% (n = 29) responded with 50% (n = 20) achieving CR and 58% (23/40) a cytogenetic response. The overall response rate and CR rate for patients with AML was 64% (n = 7) and 36% (n = 4), respectively. Patients with only TP53 mutations by next-generation sequencing had higher rates of CR (69% v 25%; P = .006). Responding patients had significant reductions in TP53 variant allele frequency and p53 expression by immunohistochemistry, with 21 (38%) achieving complete molecular remission (variant allele frequency < 5%). Median overall survival was 10.8 months with significant improvement in responding versus nonresponding patients by landmark analysis (14.6 v 7.5 months; P = .0005). Overall, 19/55 (35%) patients underwent allogeneic hematopoietic stem-cell transplant, with a median overall survival of 14.7 months. Adverse events were similar to those reported for azacitidine or eprenetapopt monotherapy, with the most common grade ≥ 3 adverse events being febrile neutropenia (33%), leukopenia (29%), and neutropenia (29%). CONCLUSION Combination treatment with eprenetapopt and azacitidine is well-tolerated yielding high rates of clinical response and molecular remissions in patients with TP53-mutant MDS and oligoblastic AML.
- Published
- 2021
- Full Text
- View/download PDF
13. A macaque clonal hematopoiesis model demonstrates expansion of TET2-disrupted clones and utility for testing interventions
- Author
-
Tae-Hoon Shin, Yifan Zhou, Shirley Chen, Stefan Cordes, Max Z. Grice, Xing Fan, Byung-Chul Lee, Aisha A. Aljanahi, So Gun Hong, Kelli L. Vaughan, Julie A. Mattison, Steven G. Kohama, Margarete A. Fabre, Naoya Uchida, Selami Demirci, Marcus A.F. Corat, Jean-Yves Métais, Katherine R. Calvo, Manuel Buscarlet, Hannah Natanson, Kathy L. McGraw, Alan F. List, Lambert Busque, John F. Tisdale, George S. Vassiliou, Kyung-Rok Yu, and Cynthia E. Dunbar
- Subjects
Inflammasomes ,Interleukin-6 ,Immunology ,Interleukin-1beta ,Cell Biology ,Hematology ,Biochemistry ,Macaca mulatta ,Hematopoiesis ,Clone Cells ,Dioxygenases ,DNA-Binding Proteins ,Young Adult ,CRISPR-Associated Protein 9 ,NLR Family, Pyrin Domain-Containing 3 Protein ,Humans ,Animals ,Clonal Hematopoiesis ,Aged - Abstract
Individuals with age-related clonal hematopoiesis (CH) are at greater risk for hematologic malignancies and cardiovascular diseases. However, predictive preclinical animal models to recapitulate the spectrum of human CH are lacking. Through error-corrected sequencing of 56 human CH/myeloid malignancy genes, we identified natural CH driver mutations in aged rhesus macaques matching genes somatically mutated in human CH, with DNMT3A mutations being the most frequent. A CH model in young adult macaques was generated via autologous transplantation of clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9–mediated gene-edited hematopoietic stem and progenitor cells (HSPCs), targeting the top human CH genes with loss-of-function (LOF) mutations. Long-term follow-up revealed reproducible and significant expansion of multiple HSPC clones with heterozygous TET2 LOF mutations, compared with minimal expansion of clones bearing other mutations. Although the blood counts of these CH macaques were normal, their bone marrows were hypercellular and myeloid-predominant. TET2-disrupted myeloid colony-forming units isolated from these animals showed a distinct hyperinflammatory gene expression profile compared with wild type. In addition, mature macrophages purified from the CH macaques showed elevated NLRP3 inflammasome activity and increased interleukin-1β (IL-1β) and IL-6 production. The model was used to test the impact of IL-6 blockage by tocilizumab, documenting a slowing of TET2-mutated expansion, suggesting that interruption of the IL-6 axis may remove the selective advantage of mutant HSPCs. These findings provide a model for examining the pathophysiology of CH and give insights into potential therapeutic interventions.
- Published
- 2022
14. TP53 mutations in myelodysplastic syndromes and secondary AML confer an immunosuppressive phenotype
- Author
-
Hsin-An Hou, Amy L. Aldrich, Amy F McLemore, Susan Geyer, Eric Padron, Kathy L. McGraw, Abhishek Dhawan, Rami S. Komrokji, Sarah Warren, John L. Cleveland, Kyle J. MacBeth, Steffen Boettcher, Alan F. List, Benjamin L. Ebert, Amy Sullivan, Jeffrey E. Lancet, Erika A. Eksioglu, Manja Meggendorfer, Najla Al Ali, Torsten Haferlach, and David A. Sallman
- Subjects
Adult ,Male ,Myeloid ,Immunology ,Gene mutation ,T-Lymphocytes, Regulatory ,Biochemistry ,medicine ,Humans ,Cytotoxic T cell ,RNA, Neoplasm ,Aged ,Aged, 80 and over ,Immunosuppression Therapy ,business.industry ,Myeloid-Derived Suppressor Cells ,Myelodysplastic syndromes ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Phenotype ,Leukemia, Myeloid, Acute ,MicroRNAs ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Mutation ,Cancer research ,Female ,Tumor Suppressor Protein p53 ,Stem cell ,business - Abstract
Somatic gene mutations are key determinants of outcome in patients with myelodysplastic syndromes (MDS) and secondary AML (sAML). In particular, patients with TP53 mutations represent a distinct molecular cohort with uniformly poor prognosis. The precise pathogenetic mechanisms underlying these inferior outcomes have not been delineated. In this study, we characterized the immunological features of the malignant clone and alterations in the immune microenvironment in patients with TP53-mutant and wild-type MDS or sAML. Notably, PDL1 expression is significantly increased in hematopoietic stem cells of patients with TP53 mutations, which is associated with MYC upregulation and marked downregulation of MYC’s negative regulator miR-34a, a p53 transcription target. Notably, patients with TP53 mutations display significantly reduced numbers of bone marrow–infiltrating OX40+ cytotoxic T cells and helper T cells, as well as decreased ICOS+ and 4-1BB+ natural killer cells. Further, highly immunosuppressive regulatory T cells (Tregs) (ie, ICOShigh/PD-1−) and myeloid-derived suppressor cells (PD-1low) are expanded in cases with TP53 mutations. Finally, a higher proportion of bone marrow–infiltrating ICOShigh/PD-1− Treg cells is a highly significant independent predictor of overall survival. We conclude that the microenvironment of TP53 mutant MDS and sAML has an immune-privileged, evasive phenotype that may be a primary driver of poor outcomes and submit that immunomodulatory therapeutic strategies may offer a benefit for this molecularly defined subpopulation.
- Published
- 2020
- Full Text
- View/download PDF
15. Comparison of mutational profiles and clinical outcomes in patients with acute myeloid leukemia with mutated RUNX1 versus acute myeloid leukemia with myelodysplasia-related changes with mutated RUNX1
- Author
-
Alan F. List, Jeffrey E. Lancet, Xiaohui Zhang, Evans Roberts, Lynn C. Moscinski, Lynh Nguyen, Ivo Abraham, David A. Sallman, Eric Padron, Dahui Qin, Diana Braswell, Seongseok Yun, Kathy L. McGraw, Ling Zhang, and Jinming Song
- Subjects
Cancer Research ,Myeloid ,business.industry ,Myeloid leukemia ,Hematology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,RUNX1 ,chemistry ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,In patient ,sense organs ,business ,neoplasms ,Acute Myeloid Leukemia with Mutated RUNX1 ,030215 immunology - Abstract
Studies comparing the prognostic role of RUNX1 mutations (RUNX1mut) in acute myeloid leukemia (AML) and acute myeloid leukemia-with myelodysplasia-related changes (AML-MRC) are limited. Our study e...
- Published
- 2020
- Full Text
- View/download PDF
16. Arginine-dependent hypusination of the eukaryotic translation initiation factor (eIF)5A drives erythroid lineage differentiation
- Author
-
Pedro Gonzalez-Menendez, Ira Phadke, Meagan E. Olive, Kathy L. McGraw, Jessica Platon, Julien Papoin, Hongxia Yan, Marie Daumur, Franciane Paul, Raghavendra Mirmira, Axel Joly, Jeremy Galtier, Anupama Narla, Guillaume Cartron, Valérie Dardalhon, Valérie S. Zimmermann, Marc Sitbon, Thomas E. Dever, Narla Mohandas, Lydie da Costa, Namrata D. Udeshi, Lionel Blanc, Sandrina Kinet, and Naomi Taylor
- Abstract
Metabolic programs contribute to hematopoietic stem and progenitor cell (HSPC) fate but it is not known whether the metabolic regulation of protein synthesis controls HSPC differentiation. We discovered that SLC7A1/CAT1-dependent arginine uptake and its catabolism to spermidine control the erythroid specification of HSPCs via activation of eukaryotic translation initiation factor 5A (eIF5A). eIF5A activity is dependent on the metabolism of spermidine to hypusine and inhibiting hypusine synthesis abrogates erythropoiesis and diverts EPO-stimulated HSPCs to a myeloid fate. Proteomic profiling reveals mitochondrial translation to be a critical target of hypusinated eIF5A and induction of mitochondrial function partially rescues erythropoiesis in the absence of hypusine. Within the hypusine network, ribosomal proteins are highly enriched and we identify defective eIF5A hypusination in erythroid pathologies caused by abnormal ribosome biogenesis. Thus, eIF5A-dependent protein synthesis is critical in the branching of erythro-myeloid differentiation and attenuated eIF5A activity characterizes ribosomal protein-linked disorders of ineffective erythropoiesis.
- Published
- 2022
- Full Text
- View/download PDF
17. Dual pyroptotic biomarkers predict erythroid response in lower-risk non-del(5q) myelodysplastic syndromes treated with lenalidomide and recombinant erythropoietin
- Author
-
Alan F. List, Jeffrey E. Lancet, Michaela Fontenay, Najla Al Ali, Chen Wang, Amy F McLemore, Olivier Kosmider, Rami S. Komrokji, Ashley A. Basiorka, Pierre Fenaux, Eric Padron, Kathy L. McGraw, and David A. Sallman
- Subjects
business.industry ,Myelodysplastic syndromes ,Hematology ,medicine.disease ,Lower risk ,Myelodysplastic Syndromes ,medicine ,Cancer research ,Erythroid response ,Chromosomes, Human, Pair 5 ,Humans ,Chromosome Deletion ,business ,Recombinant erythropoietin ,Erythropoietin ,Lenalidomide ,Biomarkers ,medicine.drug - Published
- 2021
- Full Text
- View/download PDF
18. A phase 2 trial of the oral smoothened inhibitor glasdegib in refractory myelodysplastic syndromes (MDS)
- Author
-
Kathy L. McGraw, Lisa A Nardelli, David A. Sallman, Eric Padron, Ling Zhang, Kendra Sweet, Qianxing Mo, Rami S. Komrokji, Vu H. Duong, Alan F. List, and Jeffrey E. Lancet
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Administration, Oral ,Disease ,Article ,Myeloid Neoplasm ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Aged ,Aged, 80 and over ,Salvage Therapy ,business.industry ,Phenylurea Compounds ,Myelodysplastic syndromes ,Leukemia, Myelomonocytic, Chronic ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Hedgehog signaling pathway ,Survival Rate ,Hypomethylating agent ,Drug Resistance, Neoplasm ,International Prognostic Scoring System ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Benzimidazoles ,Female ,Neoplasm Recurrence, Local ,Smoothened ,business ,Follow-Up Studies ,030215 immunology - Abstract
Hypomethylating agent (HMA) failure myelodysplastic syndrome (MDS) patients have poor outcomes and urgent need for novel therapies. Hedgehog pathway signaling upregulation plays a central role in myeloid neoplasm pathogenesis and leukemia stem cell survival. We evaluated the efficacy and safety of the smoothened inhibitor glasdegib in HMA-failure MDS (n = 35, median age 73 years). According to the International Prognostic Scoring System and the MD Anderson Global Risk Model, 54% and 77% had higher risk disease, respectively. Overall response was 6% (n = 2), and best response was marrow complete remission with hematologic improvement in both patients. Median OS and median follow-up were 10.4 and 42.8 months, respectively. Drug response/stable disease (SD) resulted in better OS than treatment failure (20.6 [95% CI, 10.4-] vs 3.9 months [95% CI, 0.7–9.1]; P < .0001). Response/SD was confirmed to be an independent covariate for improved OS (P < .0001). Grade 3 or higher infections occurred in 11% of patients (n = 4); non-hematologic toxicities were rare. Early mortality (< 30 days) occurred in 11% of patients (n = 4). Glasdegib was well tolerated among HMA-failure MDS patients, although single-agent activity was limited. SD or better resulted in notably superior OS. These results support further investigation of glasdegib, potentially in novel drug combinations, in MDS patients.
- Published
- 2019
- Full Text
- View/download PDF
19. Oxidized mitochondrial DNA released after inflammasome activation is a disease biomarker for myelodysplastic syndromes
- Author
-
Grace A Ward, Amy L. Aldrich, Eric Padron, Farnoosh Abbas-Aghababazadeh, Nghi B Lam, Benjamin S. Meyer, Olivier Kosmider, Amy F McLemore, Michaela Fontenay, Brooke L. Fridley, Erico Masala, Kathy L. McGraw, Pierre Fenaux, Javier Pinilla-Ibarz, Nicole D. Vincelette, Najla Al Ali, Joseph O. Johnson, Valeria Santini, and Alan F. List
- Subjects
Inflammasomes ,Chronic lymphocytic leukemia ,Gene mutation ,DNA, Mitochondrial ,Mice ,hemic and lymphatic diseases ,Pyroptosis ,Medicine ,Animals ,Humans ,Myeloid Neoplasia ,oxidation, myelodysplastic syndromes, inflammasome ,business.industry ,Myelodysplastic syndromes ,Hematopoietic stem cell ,Inflammasome ,Hematology ,medicine.disease ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Cancer research ,Biomarker (medicine) ,Bone marrow ,business ,Biomarkers ,medicine.drug - Abstract
Myelodysplastic syndromes (MDS) are heterogeneous hematopoietic stem cell malignancies that can phenotypically resemble other hematologic disorders. Thus, tools that may add to current diagnostic practices could aid in disease discrimination. Constitutive innate immune activation is a pathogenetic driver of ineffective hematopoiesis in MDS through Nod-like receptor protein 3 (NLRP3)–inflammasome-induced pyroptotic cell death. Oxidized mitochondrial DNA (ox-mtDNA) is released upon cytolysis, acts as a danger signal, and triggers inflammasome oligomerization via DNA sensors. By using immortalized bone marrow cells from murine models of common MDS somatic gene mutations and MDS primary samples, we demonstrate that ox-mtDNA is released upon pyroptosis. ox-mtDNA was significantly increased in MDS peripheral blood (PB) plasma compared with the plasma of healthy donors, and it was significantly higher in lower-risk MDS vs higher-risk MDS, consistent with the greater pyroptotic cell fraction in lower-risk patients. Furthermore, ox-mtDNA was significantly higher in MDS PB plasma compared with all other hematologic malignancies studied, with the exception of chronic lymphocytic leukemia (CLL). Receiver operating characteristic/area under the curve (ROC/AUC) analysis demonstrated that ox-mtDNA is a sensitive and specific biomarker for patients with MDS compared with healthy donors (AUC, 0.964), other hematologic malignancies excluding CLL (AUC, 0.893), and reactive conditions (AUC, 0.940). ox-mtDNA positively and significantly correlated with levels of known alarmins S100A9, S100A8, and apoptosis-associated speck-like protein containing caspase recruitment domain (CARD) specks, which provide an index of medullary pyroptosis. Collectively, these data indicate that quantifiable ox-mtDNA released into the extracellular space upon inflammasome activation serves as a biomarker for MDS and the magnitude of pyroptotic cell death.
- Published
- 2021
20. TFEB links MYC signaling to epigenetic control of myeloid differentiation and acute myeloid leukemia
- Author
-
Weimin Li, Gregory W Watson, Chia-Ho Cheng, Franz X. Schaub, Audrey R Freischel, Ling Cen, Hsin-An Hou, Seongseok Yun, Daniel J. Murphy, Mario R. Fernandez, Scott H. Kaufmann, Alexis Vedder, Jungwon Moon, Ling Zhang, Xiaoqing Yu, Kathy L. McGraw, Taro Hitosugi, Chunying Yang, Andrea Ballabio, Nicole D. Vincelette, John L. Cleveland, Anders Berglund, Yun, Seongseok, Vincelette, Nicole D, Yu, Xiaoqing, Watson, Gregory W, Fernandez, Mario R, Yang, Chunying, Hitosugi, Taro, Cheng, Chia-Ho, Freischel, Audrey R, Zhang, Ling, Li, Weimin, Hou, Hsinan, Schaub, Franz X, Vedder, Alexis R, Cen, Ling, Mcgraw, Kathy L, Moon, Jungwon, Murphy, Daniel J, Ballabio, Andrea, Kaufmann, Scott H, Berglund, Anders E, and Cleveland, John L
- Subjects
Myeloid ,Cellular differentiation ,Biology ,medicine.disease_cause ,Article ,Epigenesis, Genetic ,Proto-Oncogene Proteins c-myc ,hemic and lymphatic diseases ,medicine ,Humans ,Progenitor cell ,neoplasms ,Oncogene ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Myeloid leukemia ,Cell Differentiation ,General Medicine ,Isocitrate Dehydrogenase ,Haematopoiesis ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Mutation ,Cancer research ,TFEB ,Carcinogenesis ,Signal Transduction - Abstract
MYC oncoproteins regulate transcription of genes directing cell proliferation, metabolism, and tumorigenesis. A variety of alterations drive MYC expression in acute myeloid leukemia (AML), and enforced MYC expression in hematopoietic progenitors is sufficient to induce AML. Here we report that AML and myeloid progenitor cell growth and survival rely on MYC-directed suppression of Transcription Factor EB (TFEB), a master regulator of the autophagy–lysosome pathway. Notably, although originally identified as an oncogene, TFEB functions as a tumor suppressor in AML, where it provokes AML cell differentiation and death. These responses reflect TFEB control of myeloid epigenetic programs by inducing expression of isocitrate dehydrogenase-1 (IDH1) and IDH2, resulting in global hydroxylation of 5-methycytosine. Finally, activating the TFEB–IDH1/IDH2–TET2 axis is revealed as a targetable vulnerability in AML. Thus, epigenetic control by an MYC–TFEB circuit dictates myeloid cell fate and is essential for maintenance of AML. Significance: Alterations in epigenetic control are a hallmark of AML. This study establishes that a MYC–TFEB circuit controls AML differentiation and epigenetic programs by inducing IDH1/IDH2 and hydroxylation of 5-methylcytosine, that TFEB functions as a tumor suppressor in AML, and that this circuit is a targetable vulnerability in AML. See related commentary by Wu and Eisenman, p. 116.
- Published
- 2021
21. Lenalidomide induces lipid raft assembly to enhance erythropoietin receptor signaling in myelodysplastic syndrome progenitors.
- Author
-
Kathy L McGraw, Ashley A Basiorka, Joseph O Johnson, Justine Clark, Gisela Caceres, Eric Padron, Ruth Heaton, Yukiyasu Ozawa, Sheng Wei, Lubomir Sokol, and Alan F List
- Subjects
Medicine ,Science - Abstract
Anemia remains the principal management challenge for patients with lower risk Myelodysplastic Syndromes (MDS). Despite appropriate cytokine production and cellular receptor display, erythropoietin receptor (EpoR) signaling is impaired. We reported that EpoR signaling is dependent upon receptor localization within lipid raft microdomains, and that disruption of raft integrity abolishes signaling capacity. Here, we show that MDS erythroid progenitors display markedly diminished raft assembly and smaller raft aggregates compared to normal controls (p = 0.005, raft number; p = 0.023, raft size). Because lenalidomide triggers raft coalescence in T-lymphocytes promoting immune synapse formation, we assessed effects of lenalidomide on raft assembly in MDS erythroid precursors and UT7 cells. Lenalidomide treatment rapidly induced lipid raft formation accompanied by EpoR recruitment into raft fractions together with STAT5, JAK2, and Lyn kinase. The JAK2 phosphatase, CD45, a key negative regulator of EpoR signaling, was displaced from raft fractions. Lenalidomide treatment prior to Epo stimulation enhanced both JAK2 and STAT5 phosphorylation in UT7 and primary MDS erythroid progenitors, accompanied by increased STAT5 DNA binding in UT7 cells, and increased erythroid colony forming capacity in both UT7 and primary cells. Raft induction was associated with F-actin polymerization, which was blocked by Rho kinase inhibition. These data indicate that deficient raft integrity impairs EpoR signaling, and provides a novel strategy to enhance EpoR signal fidelity in non-del(5q) MDS.
- Published
- 2014
- Full Text
- View/download PDF
22. Eprenetapopt (APR-246) and Azacitidine in
- Author
-
David A, Sallman, Amy E, DeZern, Guillermo, Garcia-Manero, David P, Steensma, Gail J, Roboz, Mikkael A, Sekeres, Thomas, Cluzeau, Kendra L, Sweet, Amy, McLemore, Kathy L, McGraw, John, Puskas, Ling, Zhang, Jiqiang, Yao, Qianxing, Mo, Lisa, Nardelli, Najla H, Al Ali, Eric, Padron, Greg, Korbel, Eyal C, Attar, Hagop M, Kantarjian, Jeffrey E, Lancet, Pierre, Fenaux, Alan F, List, and Rami S, Komrokji
- Subjects
Adult ,Aged, 80 and over ,Male ,Quinuclidines ,Middle Aged ,Myelodysplastic Syndromes ,Antineoplastic Combined Chemotherapy Protocols ,Mutation ,Azacitidine ,Biomarkers, Tumor ,Humans ,Female ,Tumor Suppressor Protein p53 ,Aged - Abstract
Approximately 20% of patients withThis was a phase Ib/II study to determine the safety, recommended phase II dose, and efficacy of eprenetapopt administered in combination with azacitidine in patients withFifty-five patients (40 MDS, 11 AML, and four MDS/myeloproliferative neoplasms) with at least oneCombination treatment with eprenetapopt and azacitidine is well-tolerated yielding high rates of clinical response and molecular remissions in patients with
- Published
- 2021
23. Lenalidomide-Epoetin Alfa Versus Lenalidomide Monotherapy in Myelodysplastic Syndromes Refractory to Recombinant Erythropoietin
- Author
-
Amit Verma, Selina M. Luger, Zhuoxin Sun, Puneet S. Cheema, Jaroslaw P. Maciejewski, Martin S. Tallman, Mark R. Litzow, David F. Claxton, Jessica K. Altman, Rami S. Komrokji, John M. Bennett, Alan F. List, Kathy L. McGraw, Ryan J. Mattison, Charles A. Schiffer, Andrew S. Artz, and Timothy R. Wassenaar
- Subjects
0301 basic medicine ,Oncology ,Ineffective erythropoiesis ,Male ,Cancer Research ,medicine.medical_specialty ,Anemia ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Internal medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Recombinant erythropoietin ,Lenalidomide ,Aged ,Aged, 80 and over ,business.industry ,Myelodysplastic syndromes ,Epoetin alfa ,ORIGINAL REPORTS ,Middle Aged ,medicine.disease ,Prognosis ,Recombinant Proteins ,Epoetin Alfa ,Survival Rate ,030104 developmental biology ,Erythropoietin ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Female ,Neoplasm Recurrence, Local ,business ,medicine.drug ,Follow-Up Studies - Abstract
PURPOSE Impaired response to erythropoietin underlies ineffective erythropoiesis and anemia in myelodysplastic syndromes (MDS). We investigated whether treatment with lenalidomide (LEN), which augments erythropoietin receptor signaling in vitro, can restore and improve hemoglobin response to epoetin (EPO) alfa in patients with lower-risk, non-del(5q) MDS who have anemia that is refractory to or have low probability of benefit from treatment with recombinant erythropoietin. METHODS In a phase III, US intergroup trial, we randomly assigned patients to receive either LEN and EPO alfa or LEN alone following stratification by serum erythropoietin concentration and prior erythropoietin treatment. RESULTS A total of 195 evaluable patients were randomly assigned: 99 patients to the LEN-EPO alfa cohort and 96 to LEN alone. After four cycles of treatment, the primary end point of major erythroid response (MER) was significantly higher (28.3%) with the combination compared with LEN alone (11.5%) ( P = .004). Among 136 patients who completed 16 weeks of study treatment, 38.9% and 15.6% achieved MER, respectively ( P = .004). Additionally, minor erythroid response was achieved in 18.2% and 20.8% of patients, for an overall erythroid response rate of 46.5% versus 32.3%. Among LEN nonresponders, 38 crossed over to the addition of EPO alfa with 10 patients (26.3%) achieving a MER. Responses to the combined treatment were highly durable with a median MER duration of 23.8 months compared with 13 months with LEN alone. CONCLUSION LEN restores sensitivity to recombinant erythropoietin in growth factor–insensitive, lower-risk, non-del(5q) MDS, to yield a significantly higher rate and duration of MER compared with LEN alone (funded by the National Cancer Institute; E2905 ClinicalTrials.gov identifier: NCT02048813 ).
- Published
- 2021
24. Erythropoietin receptor signaling is membrane raft dependent.
- Author
-
Kathy L McGraw, Gwenny M Fuhler, Joseph O Johnson, Justine A Clark, Gisela C Caceres, Lubomir Sokol, and Alan F List
- Subjects
Medicine ,Science - Abstract
Upon erythropoietin (Epo) engagement, Epo-receptor (R) homodimerizes to activate JAK2 and Lyn, which phosphorylate STAT5. Although recent investigations have identified key negative regulators of Epo-R signaling, little is known about the role of membrane localization in controlling receptor signal fidelity. Here we show a critical role for membrane raft (MR) microdomains in creation of discrete signaling platforms essential for Epo-R signaling. Treatment of UT7 cells with Epo induced MR assembly and coalescence. Confocal microscopy showed that raft aggregates significantly increased after Epo stimulation (mean, 4.3±1.4(SE) vs. 25.6±3.2 aggregates/cell; p≤0.001), accompanied by a >3-fold increase in cluster size (p≤0.001). Raft fraction immunoblotting showed Epo-R translocation to MR after Epo stimulation and was confirmed by fluorescence microscopy in Epo stimulated UT7 cells and primary erythroid bursts. Receptor recruitment into MR was accompanied by incorporation of JAK2, Lyn, and STAT5 and their activated forms. Raft disruption by cholesterol depletion extinguished Epo induced Jak2, STAT5, Akt and MAPK phosphorylation in UT7 cells and erythroid progenitors. Furthermore, inhibition of the Rho GTPases Rac1 or RhoA blocked receptor recruitment into raft fractions, indicating a role for these GTPases in receptor trafficking. These data establish a critical role for MR in recruitment and assembly of Epo-R and signal intermediates into discrete membrane signaling units.
- Published
- 2012
- Full Text
- View/download PDF
25. Association of EZH2 protein expression by immunohistochemistry in myelodysplasia related neoplasms with mutation status, cytogenetics and clinical outcomes
- Author
-
Lynn C. Moscinski, Najla Al Ali, Eric Padron, Rami S. Komrokji, Alan F. List, David A. Sallman, Jinming Song, Kathy L. McGraw, Jeffrey E. Lancet, Ling Zhang, Xiaohui Zhang, and Johnny Nguyen
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Ezh2 protein ,Disease-Free Survival ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Enhancer of Zeste Homolog 2 Protein ,business.industry ,Myelodysplastic syndromes ,EZH2 ,Cytogenetics ,Hematology ,medicine.disease ,Immunohistochemistry ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Survival Rate ,030104 developmental biology ,Hematologic Neoplasms ,Myelodysplastic Syndromes ,Mutation ,Mutation (genetic algorithm) ,Cancer research ,Female ,business ,030215 immunology - Published
- 2018
- Full Text
- View/download PDF
26. Pro-inflammatory proteins S100A9 and tumor necrosis factor-α suppress erythropoietin elaboration in myelodysplastic syndromes
- Author
-
Erico Masala, Patrick Auberger, Ashley A. Basiorka, Pierre Fenaux, Brittany A. Irvine, Valeria Santini, Kathy L. McGraw, Thomas Cluzeau, Alan F. List, Sheng Wei, Jaroslaw P. Maciejewski, and Lionel Ades
- Subjects
Biology ,Peripheral blood mononuclear cell ,Article ,S100A9 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Calgranulin B ,Humans ,Erythropoiesis ,Erythropoietin ,Lenalidomide ,Tumor Necrosis Factor-alpha ,Myelodysplastic syndromes ,Hep G2 Cells ,Hematology ,medicine.disease ,Thalidomide ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Tumor necrosis factor alpha ,030215 immunology ,medicine.drug - Abstract
Accumulating evidence implicates innate immune activation in the pathobiology of myelodysplastic syndromes. A key myeloid-related inflammatory protein, S100A9, serves as a Toll-like receptor ligand regulating tumor necrosis factor-α and interleukin-1β production. The role of myelodysplastic syndrome-related inflammatory proteins in endogenous erythropoietin regulation and response to erythroid-stimulating agents or lenalidomide has not been investigated. The HepG2 hepatoma cell line was used to investigate in vitro erythropoietin elaboration. Serum samples collected from 311 patients with myelodysplastic syndrome were investigated (125 prior to treatment with erythroid-stimulating agents and 186 prior to lenalidomide therapy). Serum concentrations of S100A9, S100A8, tumor necrosis factor-α, interleukin-1β and erythropoietin were analyzed by enzyme-linked immunosorbent assay. Using erythropoietin-producing HepG2 cells, we show that S100A9, tumor necrosis factor-α and interleukin-1β suppress transcription and cellular elaboration of erythropoietin. Pre-incubation with lenalidomide significantly diminished suppression of erythropoietin production by S100A9 or tumor necrosis factor-α. Moreover, in peripheral blood mononuclear cells from patients with myelodysplastic syndromes, lenalidomide significantly reduced steady-state S100A9 generation (P=0.01) and lipopolysaccharide-induced tumor necrosis factor-α elaboration (P=0.002). Enzyme-linked immunosorbent assays of serum from 316 patients with non-del(5q) myelodysplastic syndromes demonstrated a significant inverse correlation between tumor necrosis factor-α and erythropoietin concentrations (P=0.006), and between S100A9 and erythropoietin (P=0.01). Moreover, baseline serum tumor necrosis factor-α concentration was significantly higher in responders to erythroid-stimulating agents (P=0.03), whereas lenalidomide responders had significantly lower tumor necrosis factor-α and higher S100A9 serum concentrations (P=0.03). These findings suggest that S100A9 and its nuclear factor-κB transcriptional target, tumor necrosis factor-α, directly suppress erythropoietin elaboration in myelodysplastic syndromes. These cytokines may serve as rational biomarkers of response to lenalidomide and erythroid-stimulating agent treatments. Therapeutic strategies that either neutralize or suppress S100A9 may improve erythropoiesis in patients with myelodysplastic syndromes.
- Published
- 2017
- Full Text
- View/download PDF
27. Tfeb Links MYC Signaling to Epigenetic Control of Acute Myeloid Leukemia Cell Death and Differentiation
- Author
-
Scott H. Kaufmann, Mario R. Fernandez, Audrey R Freischel, Taro Hitosugi, Franz X. Schaub, Ling Zhang, Alexis Vedder, Hsin-An Hou, Seongseok Yun, Kathy L. McGraw, Chia-Ho Cheng, Ling Cen, Chunying Yang, Nicole D. Vincelette, Daniel J. Murphy, Xiaoqing Yu, Andrea Ballabio, John L. Cleveland, Anders Berglund, Gregory W Watson, and Weimin Li
- Subjects
Programmed cell death ,Immunology ,Cancer research ,TFEB ,Myeloid leukemia ,Cell Biology ,Hematology ,Epigenetics ,Biology ,Biochemistry - Abstract
MYC gene amplification and somatic mutations are frequent in both adult and pediatric AML although how MYC drives and contributes to the development and maintenance of AML has not been resolved. Transcription factor EB (TFEB) is a master regulator of genes that control autophagy and lysosome biogenesis, a central catabolic recycling pathway that regulates cell survival. Given the oncogenic effects of MYC in AML and that the induction of autophagy compromises AML cell growth and survival, we tested if the oncogenic effect of MYC depends on its suppression of TFEB transcription programs in AML. In support of this hypothesis, inducible MYC expression in K562 and THP-1 leukemia cells was sufficient to suppress expression of TFEB and its target genes. Further and conversely, MYC knockdown in NB4 AML cells provoked increased expression of TFEB mRNA and protein, as well as increased expression of TFEB target genes. Notably, dose response studies demonstrated that expression of TFEBS211A, constitutively nuclear form of TFEB that is refractory to control by mTORC1 signaling, dramatically impairs proliferation of HL60, OCI-AML2 and OCI-AML3 AML cells. In addition, induction of TFEBS211A provoked rampant apoptosis. Of important, overexpression of TFEBS211A in HL-60 and OCI-AML3 cells was also sufficient to promote monocytic and granulocytic differentiation, as judged by morphological changes and the acquisition of mature monocytic and granulocytic markers including CD11b, Gr1, and CD15. To identify TFEB targets that might contribute to myeloid/granulocytic differentiation, we performed RNA-seq analysis of HL60 leukemia cells engineered to inducibly express the TFEBS211A transgene. Using a cut-off of fold change>4 with q Surprisingly, among genes induced by TFEB in HL60 cells is IDH1, which catalyzes the production of α-ketoglutarate (α-KG), a required substrate of the TET family of dioxygenases that convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). In particular, TFEBS211A expression provoked increases in levels of α-KG and significant increases in global levels of 5hmC in genomic DNA of HL60 leukemia cells both ex vivo and in vivo. Furthermore, TFEB-mediated induction of IDH1/2 mRNA and protein, and of IDH1 promoter activity, was antagonized by inducible expression of MYC. To assess the global effects of TFEB on 5mC/5hmC landscapes, we performed paired reduced representation bisulfite (BS)- and oxidation bisulfite (oxBS)-sequencing. As predicted, TFEBS211A induced both loss and gains of 5mC, but there were more losses (n = 722) than gains (n = 459) across all 22 chromosomes. Quite remarkably, and consistent with TFEB-provoked increases of 5hmC signals, TFEBS211A exclusively induced 5hmC gains (in a total of 863 genes), and 37% and 36% of these 5hmC gains occurred in promoter regions and CpG islands, and across all 22 chromosomes. Comparison of BS- and oxBS-seq versus RNA-seq analyses of HL60 cells expressing TFEB revealed significant changes in mRNA levels and concomitant differential changes in 5mC and 5hmC marks in KLF4, KLF6, STAT3, TP73, andFOXO1 that have pivotal roles in controlling myeloid cell differentiation and death. Collectively, these findings demonstrate that MYC suppresses TFEB expression and function in AML cells, and that TFEB functions as a tumor suppressor that provokes AML cell differentiation and death. Strikingly, these responses rely on epigenetic control, where TFEB directly induces the transcription of IDH1 and IDH2 to provoke global hydroxylation of 5-methylcytosine and the expression of genes that drive terminal differentiation and apoptosis. Thus, a MYC-TFEB-IDH1/2-TET2 circuit controls AML cell fate. Disclosures Murphy: Merck: Research Funding; Puma Biotech: Research Funding. Ballabio:CASMA Therapeutics: Other: Co-Founder. Kaufmann:Takeda Pharmaceuticals: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
28. Non-del(5q) myelodysplastic syndromes–associated loci detected by SNP-array genome-wide association meta-analysis
- Author
-
Chimène Moreilhon, Alan F. List, Peter A. Kanetsky, Mar Mallo, Azim M Mohamedali, Bartlomiej P Przychodzen, Sophie Raynaud, Francesc Solé, Jacqueline Boultwood, Thomas Cluzeau, Hwei-Fang Tien, Björn Nilsson, Kathy L. McGraw, Pierre Fenaux, Y. Ann Chen, Leonor Arenillas, David A. Sallman, Eric Padron, Ghulam J. Mufti, Chia-Ho Cheng, Jaroslaw P. Maciejewski, Giulio Genovese, Lionel Ades, Lubomir Sokol, Andrea Pellagatti, Benjamin L. Ebert, and Hsin-An Hou
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Quantitative Trait Loci ,Single-nucleotide polymorphism ,Genome-wide association study ,Biology ,Genoma humà ,Polymorphism, Single Nucleotide ,Germline ,Germline mutation ,hemic and lymphatic diseases ,medicine ,Humans ,Genetic Predisposition to Disease ,Cromosomes humans ,Polimorfisme cromosòmic ,Myeloid Neoplasia ,Myelodysplastic syndromes ,Hematology ,Genomics ,medicine.disease ,medicine.anatomical_structure ,Gene Expression Regulation ,Myelodysplastic Syndromes ,Immunology ,Chromosomes, Human, Pair 5 ,Bone marrow ,Chromosome Deletion ,SNP array ,Genome-Wide Association Study - Abstract
Myelodysplastic syndromes (MDS) are hematopoietic stem cell malignancies. Known predisposing factors to adult MDS include rare germline mutations, cytotoxic therapy, age-related clonal hematopoiesis, and autoimmune or chronic inflammatory disorders. To date, no published studies characterizing MDS-associated germline susceptibility polymorphisms exist. We performed a genome-wide association study of 2 sample sets (555 MDS cases vs 2964 control subjects; 352 MDS cases vs 2640 control subjects) in non-del(5q) MDS cases of European genomic ancestry. Meta-analysis identified 8 MDS-associated loci at 1q31.1 (PLA2G4A), 3p14.1 (FAM19A4), 5q21.3 (EFNA5), 6p21.33, 10q23.1 (GRID1), 12q24.32, 15q26.1, and 20q13.12 (EYA2) that approached genome-wide significance. Gene expression for 5 loci that mapped within or near genes was significantly upregulated in MDS bone marrow cells compared with those of control subjects (P < .01). Higher PLA2G4A expression and lower EYA2 expression were associated with poorer overall survival (P = .039 and P = .037, respectively). Higher PLA2G4A expression is associated with mutations in NRAS (P < .001), RUNX1 (P = .012), ASXL1 (P = .007), and EZH2 (P = .038), all of which are known to contribute to MDS development. EYA2 expression was an independently favorable risk factor irrespective of age, sex, and Revised International Scoring System score (relative risk, 0.67; P = .048). Notably, these genes have regulatory roles in innate immunity, a critical driver of MDS pathogenesis. EYA2 overexpression induced innate immune activation, whereas EYA2 inhibition restored colony-forming potential in primary MDS cells indicative of hematopoietic restoration and possible clinical relevance. In conclusion, among 8 suggestive MDS-associated loci, 5 map to genes upregulated in MDS with functional roles in innate immunity and potential biological relevance to MDS.
- Published
- 2019
29. Cytokine-Regulated Phosphorylation and Activation of TET2 by JAK2 in Hematopoiesis
- Author
-
Sriram Sundaravel, Tushar D. Bhagat, Ji Nie, Yogen Saunthararajah, Michael J. Thirman, Kith Pradhan, James J. Bieker, Peng Ji, Sangeeta Nischal, Alison R. Moliterno, Kathy L. McGraw, Michael R. Bishop, Amit Verma, Xiaorong Gu, Alan F. List, Hui Liu, Andrew S. Artz, John Z. Cao, Amittha Wickrema, Ulrich Steidl, Lucy A. Godley, Ross L. Levine, Chuan He, Sanchari Bhattacharyya, Wen-Liang Kuo, and Jong Jin Jeong
- Subjects
0301 basic medicine ,DNA Hydroxymethylation ,Transcriptional Activation ,KLF1 ,Chromatin remodeling ,Article ,Dioxygenases ,03 medical and health sciences ,0302 clinical medicine ,Myeloproliferative Disorders ,Proto-Oncogene Proteins ,Humans ,Epigenetics ,Phosphorylation ,Chemistry ,GATA1 ,Janus Kinase 2 ,Hematopoietic Stem Cells ,Cell biology ,Hematopoiesis ,DNA-Binding Proteins ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,Cytokines ,Biomarkers - Abstract
Even though the Ten-eleven translocation (TET) enzymes catalyze the generation of 5-hydroxymethylcytosines required for lineage commitment and subsequent differentiation of stem cells into erythroid cells, the mechanisms that link extracellular signals to TET activation and DNA hydroxymethylation are unknown. We demonstrate that hematopoietic cytokines phosphorylate TET2, leading to its activation in erythroid progenitors. Specifically, cytokine receptor–associated JAK2 phosphorylates TET2 at tyrosines 1939 and 1964. Phosphorylated TET2 interacts with the erythroid transcription factor KLF1, and this interaction with TET2 is increased upon exposure to erythropoietin. The activating JAK2V617F mutation seen in myeloproliferative disease patient samples and in mouse models is associated with increased TET activity and cytosine hydroxymethylation as well as genome-wide loss of cytosine methylation. These epigenetic and functional changes are also associated with increased expression of several oncogenic transcripts. Thus, we demonstrate that JAK2-mediated TET2 phosphorylation provides a mechanistic link between extracellular signals and epigenetic changes during hematopoiesis. Significance: Identification of TET2 phosphorylation and activation by cytokine-stimulated JAK2 links extracellular signals to chromatin remodeling during hematopoietic differentiation. This provides potential avenues to regulate TET2 function in the context of myeloproliferative disorders and myelodysplastic syndromes associated with the JAK2V617F-activating mutation. This article is highlighted in the In This Issue feature, p. 681
- Published
- 2019
- Full Text
- View/download PDF
30. Loss of Function of DOCK4 in Myelodysplastic Syndromes Stem Cells is Restored by Inhibitors of DOCK4 Signaling Networks
- Author
-
Gaurav Choudhary, Shanisha Gordon-Mitchell, Amit Verma, Amittha Wickrema, Sriram Sundaravel, Sandeep Gurbuxani, Kathy L. McGraw, Hui Liu, Jong Jin Jeong, Wen-Liang Kuo, Tushar D. Bhagat, and Alan F. List
- Subjects
0301 basic medicine ,Cancer Research ,PTPN6 ,Antineoplastic Agents ,Protein tyrosine phosphatase ,Biology ,Models, Biological ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,LYN ,Cell Movement ,Loss of Function Mutation ,hemic and lymphatic diseases ,Humans ,INPP5D ,Phosphorylation ,Regulation of gene expression ,Protein Tyrosine Phosphatase, Non-Receptor Type 6 ,GTPase-Activating Proteins ,Tyrosine phosphorylation ,Hematopoietic Stem Cells ,Cell biology ,Gene Expression Regulation, Neoplastic ,Haematopoiesis ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases ,Protein Binding ,Signal Transduction - Abstract
Purpose: Myelodysplastic syndromes (MDS) with deletion of chromosome 7q/7 [-7/(del)7q MDS] is associated with worse outcomes and needs novel insights into pathogenesis. Reduced expression of signaling protein dedicator of cytokinesis 4 (DOCK4) in patients with -7/(del)7q MDS leads to a block in hematopoietic stem cell (HSC) differentiation. Identification of targetable signaling networks downstream of DOCK4 will provide means to restore hematopoietic differentiation in MDS. Experimental Design: We utilized phosphoproteomics approaches to identify signaling proteins perturbed as a result of reduced expression of DOCK4 in human HSCs and tested their functional significance in primary model systems. Results: We demonstrate that reduced levels of DOCK4 lead to increased global tyrosine phosphorylation of proteins in primary human HSCs. LYN kinase and phosphatases INPP5D (SHIP1) and PTPN6 (SHP1) displayed greatest levels of tyrosine phosphorylation when DOCK4 expression levels were reduced using DOCK4-specific siRNA. Our data also found that increased phosphorylation of SHIP1 and SHP1 phosphatases were due to LYN kinase targeting these phosphatases as substrates. Increased migration and impediment of HSC differentiation were consequences of these signaling alterations. Pharmacologic inhibition of SHP1 reversed these functional aberrations in HSCs expressing low DOCK4 levels. In addition, differentiation block seen in DOCK4 haplo-insufficient [-7/(del)7q] MDS was rescued by inhibition of SHP1 phosphatase. Conclusions: LYN kinase and phosphatases SHP1 and SHIP1 are perturbed when DOCK4 expression levels are low. Inhibition of SHP1 promotes erythroid differentiation in healthy HSCs and in -7/(del)7q MDS samples with low DOCK4 expression. Inhibitors of LYN, SHP1 and SHIP1 also abrogated increased migratory properties in HSCs expressing reduced levels of DOCK4.
- Published
- 2019
31. Using tissue microarray to detect inflammasome signaling components that contribute to the pathogenesis of myelodysplastic syndrome
- Author
-
Jeffrey E. Lancet, Seongseok Yun, Kathy L. McGraw, Rohit Sharma, Rami S. Komrokji, Ling Zhang, Tony Kurian, and Sanjay Bridgelall
- Subjects
Ineffective Hematopoiesis ,Cancer Research ,Cytopenia ,Tissue microarray ,business.industry ,Myelodysplastic syndromes ,Myeloid leukemia ,Inflammasome ,medicine.disease ,Pathogenesis ,Oncology ,hemic and lymphatic diseases ,medicine ,Cancer research ,business ,medicine.drug - Abstract
7044 Background: Myelodysplastic syndromes (MDS) are characterized by aberrant maturation, ineffective hematopoiesis, cytopenia, and progression to acute myeloid leukemia. MDS pathogenesis is multifactorial and potentially linked to constitutive innate immune stimulation converging upon the NLRP3 inflammasome to induce pyroptosis, a caspase-1 dependent cell death. Inflammasome assembly is initiated by both cell-extrinsic stimuli including S100A9, the TLR4 and CD33 ligand, and cell-intrinsic danger signals licensing caspase-1 which activates IL1b and beta-catenin resulting in cell death and cellular proliferation leading to maturation and differentiation blocks. Further, EYA2 has been suggested to be an inflammasome activator, whereas cPLA2 has been suggested to be an inhibitor. The purpose of this study is to determine whether immunohistochemistry (IHC) may be utilized to assess expression of inflammasome components. Methods: An IRB protocol was approved prior to initiating this study. We retrospectively identified 43 low risk MDS patients. A tissue microarray (TMA) was constructed utilizing MDS bone marrow biopsy samples (2-3 representative cores per sample). IL-1, S100A9, EYA2, cPLA2, beta-catenin, and TLR4 expression were assessed by IHC after validation of each antibody. IHC expression was scored independently by two hematopathologists by calculating scores (product of staining intensity x percent expression). IHC expression was compared using Spearman correlation estimate. Demographic and clinical data were collected and correlated with IHC expression using Kruskal-Wallis test, Spearman correlation, and Logrank test. Results: Patients were median 72 years of age, 67% men and included 47% MDS-MLD, 35% MDS-RS, 14% MDS-SLD, 2% MDS del5q and 2% MDS-U. IL-1 expression correlated with beta-catenin expression, r = 0.42, 95% CI 0.115 to 0.658 (p = 0.007). There was a trend towards significance between IL-1 and cPLA2, r = 0.30 (p = 0.067); S100A9 and cPLA2, r = 0.31 (p = 0.052); and S100A9 and EYA2, r = 0.31 (p = 0.057). Percentage EYA2 expression correlated with blast count, r = 0.425 (p = 0.008). The IHC expression of these antigens did not correlate with WHO MDS subclassification, IPSS, R-IPSS, disease progression, or survival (p > 0.05). Conclusions: IHC staining of inflammasome activators using TMA may allow better characterization of molecular pathways contributing the MDS pathogenesis. A correlation was seen between expression of antigens known to be increased downstream of NLRP3 inflammasome activation. Furthermore, increased expression of EYA2 correlated with blast count. A future study will compare expression patterns between normal, low risk MDS and high risk MDS samples and correlate these findings with clinical outcome data to further elucidate the pathogenesis of MDS and identify potential targetable markers for novel therapeutic strategies.
- Published
- 2021
- Full Text
- View/download PDF
32. The NLRP3 inflammasome functions as a driver of the myelodysplastic syndrome phenotype
- Author
-
David A. Sallman, Avril A. B. Robertson, Erika A. Eksioglu, John L. Cleveland, Rami S. Komrokji, Thomas Cluzeau, Xianghong Chen, Ling Zhang, Mark E. Cooper, Luke A. J. O'Neill, Sheng Wei, Lubomir Sokol, Joseph O. Johnson, Kathy L. McGraw, Rebecca C. Coll, Ashley A. Basiorka, Qing Zhang, Alan F. List, Brittany A. Irvine, and Eric Padron
- Subjects
0301 basic medicine ,Inflammasomes ,Somatic cell ,Immunology ,Apoptosis ,Mice, Transgenic ,Gene mutation ,Biochemistry ,Ion Channels ,Colony-Forming Units Assay ,03 medical and health sciences ,NLR Family, Pyrin Domain-Containing 3 Protein ,Pyroptosis ,medicine ,Animals ,Calgranulin B ,Humans ,Progenitor cell ,beta Catenin ,Cell Size ,Ineffective Hematopoiesis ,Myeloid Neoplasia ,NADPH oxidase ,biology ,Caspase 1 ,NADPH Oxidases ,Inflammasome ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Hematopoiesis ,Haematopoiesis ,Phenotype ,030104 developmental biology ,Myelodysplastic Syndromes ,Mutation ,biology.protein ,Cancer research ,Reactive Oxygen Species ,Ion Channel Gating ,medicine.drug - Abstract
Despite genetic heterogeneity, myelodysplastic syndromes (MDSs) share features of cytological dysplasia and ineffective hematopoiesis. We report that a hallmark of MDSs is activation of the NLRP3 inflammasome, which drives clonal expansion and pyroptotic cell death. Independent of genotype, MDS hematopoietic stem and progenitor cells (HSPCs) overexpress inflammasome proteins and manifest activated NLRP3 complexes that direct activation of caspase-1, generation of interleukin-1β (IL-1β) and IL-18, and pyroptotic cell death. Mechanistically, pyroptosis is triggered by the alarmin S100A9 that is found in excess in MDS HSPCs and bone marrow plasma. Further, like somatic gene mutations, S100A9-induced signaling activates NADPH oxidase (NOX), increasing levels of reactive oxygen species (ROS) that initiate cation influx, cell swelling, and β-catenin activation. Notably, knockdown of NLRP3 or caspase-1, neutralization of S100A9, and pharmacologic inhibition of NLRP3 or NOX suppress pyroptosis, ROS generation, and nuclear β-catenin in MDSs and are sufficient to restore effective hematopoiesis. Thus, alarmins and founder gene mutations in MDSs license a common redox-sensitive inflammasome circuit, which suggests new avenues for therapeutic intervention.
- Published
- 2016
- Full Text
- View/download PDF
33. Molecular predictors of response in patients with myeloid neoplasms treated with lenalidomide
- Author
-
Aziz Nazha, Michael J. Clemente, Vera Adema, Eiju Negoro, Bartlomiej P Przychodzen, Tomas Radivoyevitch, Francesc Sole, Valeria Santini, Jaroslaw P. Maciejewski, Alan F. List, Chantana Polprasert, Hideki Makishima, Kathy L. McGraw, Naoko Hosono, Mikkael A. Sekeres, and Cassandra M. Hirsch
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Article ,Cohort Studies ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Hematology ,Anesthesiology and Pain Medicine ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Immunologic Factors ,Lenalidomide ,Aged ,Aged, 80 and over ,business.industry ,Myelodysplastic syndromes ,Middle Aged ,medicine.disease ,3. Good health ,Lymphoma ,Thalidomide ,stomatognathic diseases ,Haematopoiesis ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Mutation ,Chromosomes, Human, Pair 5 ,Female ,business ,medicine.drug - Abstract
Molecular predictors of response in patients with myeloid neoplasms treated with lenalidomide
- Published
- 2016
34. Immunohistochemical pattern of p53 is a measure of TP53 mutation burden and adverse clinical outcome in myelodysplastic syndromes and secondary acute myeloid leukemia
- Author
-
Lynn C. Moscinski, Eric Padron, Johnny Nguyen, Kathy L. McGraw, Rami S. Komrokji, Ling Zhang, Alan F. List, David A. Sallman, Jeffrey E. Lancet, and Najla Al Ali
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Gene mutation ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,hemic and lymphatic diseases ,Myeloblast ,Internal medicine ,medicine ,Humans ,Secondary Acute Myeloid Leukemia ,Online Only Articles ,TP53 Gene Mutation ,Allele frequency ,Survival analysis ,Aged ,Retrospective Studies ,Myelodysplastic syndromes ,Myeloid leukemia ,Neoplasms, Second Primary ,Hematology ,Prognosis ,medicine.disease ,Immunohistochemistry ,Survival Analysis ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Cytogenetic Analysis ,Immunology ,Female ,Tumor Suppressor Protein p53 ,030215 immunology - Abstract
Myelodysplastic syndromes (MDS) are genetically diverse malignancies with peripheral cytopenias, dysplastic hematopoiesis, and increased risk for acute myeloid leukemia (AML) transformation. Recent investigations indicate that somatic, myeloid-specific gene mutations refine clinical staging to alter estimates of overall survival (OS), and should be included in current risk stratification models.1 Hence, the identification of these mutations and corresponding protein expression levels has increasing clinical utility. TP53 mutations are found in 5–10% of MDS patients, are enriched in patients with isolated del(5q), complex cytogenetics, or MDS with fibrosis (MDS-F), and are associated with an overall worse prognosis.1–5 Next-generation sequencing (NGS) is a valuable ancillary tool, however, the technology may not be economically feasible for routine community use. Alternatively, immunohistochemistry (IHC) is fast, reproducible, and cost effective for routine laboratory use. In this study, we explore the relationship between p53 expression and TP53 gene mutation in MDS and acute myeloid leukemia with myelodysplasia-related changes (AML-MRC). Additionally, we investigate correlations between p53 expression and clinical characteristics, including TP53 mutation variant allele frequency (VAF), myeloblast percentage, cytogenetic characteristics and outcome.
- Published
- 2016
- Full Text
- View/download PDF
35. TP53 and MDM2 single nucleotide polymorphisms influence survival in non-del(5q) myelodysplastic syndromes
- Author
-
Dana E. Rollison, Lubomir Sokol, David A. Sallman, Jaroslaw P. Maciejewski, Pearlie K. Epling-Burnette, Ling Zhang, Ashley A. Basiorka, Mar Mallo, Thomas Cluzeau, Alan F. List, Brittany A. Irvine, Kathy L. McGraw, and Francesc Solé
- Subjects
Adult ,Male ,Survival ,Genotype ,Myelodysplastic syndromes ,Single-nucleotide polymorphism ,Kaplan-Meier Estimate ,Biology ,Bioinformatics ,survival ,Polymorphism, Single Nucleotide ,Disease-Free Survival ,03 medical and health sciences ,symbols.namesake ,single nucleotide polymorphisms ,0302 clinical medicine ,MDM2 ,medicine ,SNP ,Humans ,TP53 ,030304 developmental biology ,Aged ,Proportional Hazards Models ,Sanger sequencing ,Aged, 80 and over ,0303 health sciences ,Univariate analysis ,Proportional hazards model ,Proto-Oncogene Proteins c-mdm2 ,Single nucleotide polymorphisms ,Middle Aged ,medicine.disease ,Prognosis ,myelodysplastic syndromes ,3. Good health ,Oncology ,030220 oncology & carcinogenesis ,symbols ,biology.protein ,Mdm2 ,Female ,Tumor Suppressor Protein p53 ,Research Paper - Abstract
Altres ajuts: National Cancer Institute/National Institute of Health (5 R01CA131076-04); Molecular Genomics Facility (P30-CA076292); Sociedad Española Hematología y Hemoterapia (2014 SGR225); Fundació Internacional Josep Carreras i Celgene Spain. RTICC/RD12/0036/0044 Abstract:P53 is a key regulator of many cellular processes and is negatively regulated by the human homolog of murine double minute-2 (MDM2) E3 ubiquitin ligase. Single nucleotide polymorphisms (SNPs) of either gene alone, and in combination, are linked to cancer susceptibility, disease progression, and therapy response. We analyzed the interaction of TP53 R72P and MDM2 SNP309 SNPs in relationship to outcome in patients with myelodysplastic syndromes (MDS). Sanger sequencing was performed on DNA isolated from 208 MDS cases. Utilizing a novel functional SNP scoring system ranging from +2 to -2 based on predicted p53 activity, we found statistically significant differences in overall survival (OS) (p = 0.02) and progression-free survival (PFS) (p = 0.02) in non-del(5q) MDS patients with low functional scores. In univariate analysis, only IPSS and the functional SNP score predicted OS and PFS in non-del(5q) patients. In multivariate analysis, the functional SNP score was independent of IPSS for OS and PFS. These data underscore the importance of TP53 R72P and MDM2 SNP309 SNPs in MDS, and provide a novel scoring system independent of IPSS that is predictive for disease outcome.
- Published
- 2015
36. Assessment of ASC specks as a putative biomarker of pyroptosis in myelodysplastic syndromes: an observational cohort study
- Author
-
Michaela Fontenay, Sheng Wei, Brooke L. Fridley, Erika A. Eksioglu, Grace A Ward, Kathy L. McGraw, Amy F McLemore, Najla Al Ali, David A. Sallman, Olivier Kosmider, Erico Masala, Lubomir Sokol, Rami S. Komrokji, Eric Padron, Farnoosh Abbas-Aghababazadeh, Pierre Fenaux, Ashley A. Basiorka, Nicole D. Vincelette, Alan F. List, Valeria Santini, and Javier Pinilla-Ibarz
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Article ,Cohort Studies ,03 medical and health sciences ,Aged ,Biomarkers ,CARD Signaling Adaptor Proteins ,Case-Control Studies ,Female ,Humans ,Myelodysplastic Syndromes ,Pyroptosis ,0302 clinical medicine ,Internal medicine ,medicine ,business.industry ,Myelodysplastic syndromes ,Area under the curve ,Case-control study ,Cancer ,PYCARD ,Inflammasome ,Hematology ,medicine.disease ,3. Good health ,030104 developmental biology ,Cohort ,Biomarker (medicine) ,business ,030215 immunology ,medicine.drug ,Signal Transduction - Abstract
Summary Background NLRP3 inflammasome-directed pyroptotic cell death drives ineffective haemopoiesis in myelodysplastic syndromes. During inflammasome assembly, the apoptosis-associated speck-like protein containing a CARD (PYCARD, commonly known as ASC) adaptor protein polymerises into large, filamentous clusters termed ASC specks that are released upon cytolysis. Specks are resistant to proteolytic degradation because of their prion-like structure, and therefore might serve as a biomarker for pyroptotic cell death in myelodysplastic syndromes. Methods This observational cohort study was done at the H Lee Moffitt Cancer Center (Tampa, FL, USA). Patients with myelodysplastic syndromes, healthy controls, and patients with non-myelodysplastic syndrome haematological cancers or type 2 diabetes were recruited. We used confocal and electron microscopy to visualise, and flow cytometry to quantify, ASC specks in peripheral blood and bone marrow plasma samples. Speck percentages were compared by t test or ANOVA, correlations were assessed by Spearman's rank correlation coefficient, and biomarker efficiency was assessed by receiver operating characteristics and area under the curve (AUC) analysis. Findings Between Jan 1, 2005, and Jan 12, 2017, we obtained samples from 177 patients with myelodysplastic syndromes and 29 healthy controls for the discovery cohort, and 113 patients with myelodysplastic syndromes and 31 healthy controls for the validation cohort. We also obtained samples from 22 patients with del(5q) myelodysplastic syndromes, 230 patients with non-myelodysplastic syndrome haematological cancers and 23 patients with type 2 diabetes. After adjustment for glucose concentration, the log10-transformed mean percentage of peripheral blood plasma-derived ASC specks was significantly higher in the 177 patients with myelodysplastic syndromes versus the 29 age-matched, healthy donors (−0·41 [SD 0·49] vs −0·67 [0·59], p=0·034). The percentages of ASC specks in samples from patients with myelodysplastic syndromes were significantly greater than those in samples from individuals with every other haematological cancer studied (all p Interpretation Our results underscore the pathobiological relevance of ASC specks and suggest that ASC specks are a sensitive and specific candidate plasma biomarker that provides an index of medullary pyroptotic cell death and ineffective haemopoiesis in patients with myelodysplastic syndromes. Funding T32 Training Grant (NIH/NCI 5T32 CA115308–08), Edward P Evans Foundation, The Taub Foundation Grants Program, the Flow Cytometry, Analytic Microscopy, and Tissue Core Facilities at the H Lee Moffitt Cancer Center and Research Institute, a National Cancer Institute-designated Comprehensive Cancer Center (P30-CA076292).
- Published
- 2018
37. Erythropoietin Receptor Signaling and Lipid Rafts
- Author
-
Kathy L. McGraw and Alan F. List
- Subjects
0301 basic medicine ,Article ,03 medical and health sciences ,0302 clinical medicine ,Membrane Microdomains ,hemic and lymphatic diseases ,medicine ,Receptors, Erythropoietin ,Animals ,Humans ,Receptor ,Transcription factor ,Lipid raft ,Erythropoietin ,STAT5 ,biology ,Sphingolipid ,Erythropoietin receptor ,Cell biology ,Red blood cell ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,biology.protein ,Phosphorylation ,lipids (amino acids, peptides, and proteins) ,Signal Transduction - Abstract
Erythropoiesis is tightly regulated by the growth factor erythropoietin (Epo). Signal activation begins when Epo engages its cognate receptor, Epo-R, triggering receptor homodimerization, and recruitment of signaling intermediates including Jak2 that phosphorylates both the receptor cytoplasmic tail and downstream effectors including the transcription factor, STAT5. Transcription factors subsequently activate transcription of prosurvival and prodifferentiation genes responsible for red blood cell production. The fidelity of Epo-R signaling is dependent upon residence within detergent insoluble membrane lipid raft fractions. Lipid rafts are membrane microdomains that serve as signaling scaffolds composed of densely packed sphingolipids and cholesterol where receptors and intermediate signaling proteins are recruited and interact to execute stimuli. Disruption of lipid rafts is detrimental to Epo signaling, a phenomenon that may be utilized to design novel therapeutics for conditions in which Epo signaling is deficient. Here, we review the Epo signaling cascade, particularly, as it relates to localization and dependence on lipid rafts, and discuss considerations for novel therapeutic design.
- Published
- 2017
38. Phase 2 Results of APR-246 and Azacitidine (AZA) in Patients with TP53 mutant Myelodysplastic Syndromes (MDS) and Oligoblastic Acute Myeloid Leukemia (AML)
- Author
-
Rami S. Komrokji, Hagop M. Kantarjian, Najla Al Ali, Amy E. DeZern, John Puskas, Ling Zhang, Alan F. List, Jeffrey E. Lancet, David A. Sallman, Thomas Cluzeau, Kendra Sweet, Guillermo Garcia-Manero, Eyal C. Attar, Amy F McLemore, David P. Steensma, Gail J. Roboz, Qianxing Mo, Pierre Fenaux, Greg Korbel, Jiqiang Yao, Lisa A Nardelli, Eric Padron, Mikkael A. Sekeres, and Kathy L. McGraw
- Subjects
Cytopenia ,Leukopenia ,business.industry ,medicine.medical_treatment ,Myelodysplastic syndromes ,Immunology ,Mutant ,Azacitidine ,Myeloid leukemia ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Cancer research ,medicine ,medicine.symptom ,business ,Febrile neutropenia ,medicine.drug - Abstract
Introduction: TP53 gene mutations (mTP53), found in up to 20% of MDS or AML pts and 30-40% of therapy-related (TR) MDS/AML cases, represent a distinct molecular cohort with poor outcomes. Hypomethylating agents (HMA) are the standard of care with CR rates of ~20% and median OS of 7-8 months. APR-246 is a novel, first-in-class small molecule that selectively induces apoptosis in mTP53 cancer cells via thermodynamic stabilization of the p53 protein and shifting equilibrium toward the wild-type conformation. We previously reported the Phase 1b results of APR-246+AZA with no DLTs, transcriptional activation of p53 targets and high response rates, identifying a Phase 2 (P2) dose of 4500mg days 1-4 (Sallman et al., ASH 2018). We report herein the planned, completed phase 2 results. Methods: This is a multicenter Phase 1b/2 trial of APR-246+AZA in HMA-naïve mTP53 higher risk MDS, MDS/MPN and oligoblastic AML (≤ 30% blasts) pts (NCT03072043). P2 pts received APR-246 4500mg IV (days 1-4) + AZA 75 mg/m2 SC/IV x 7 days (days 4-10 or 4-5 and 8-12) in 28 day cycles. Primary objective was CR rate by International Working Group (IWG) 2006 criteria. Secondary objectives included ORR, OS, outcome following allogeneic hematopoietic stem cell transplant (allo-HSCT), and both next generation sequencing (NGS) and p53 immunohistochemistry (IHC) to monitor clonal suppression and remission depth as prognostic covariates. For minimal residual disease (MRD) analysis, a custom target-capture NGS assay was developed using unique molecular Identifiers for error correction with a 0.1% limit of detection. Results: As of July 15, 2019, 55 pts were enrolled (6 P1; 49 P2) with a median age 66 years (34-85; 47% male). By WHO, 40 pts had MDS, 11 AML-MRC and 4 CMML/MDS-MPN; 85% had complex cytogenetics and 33% TR-MDS/AML. All pts had higher risk disease by IPSS-R (7% Intermediate, 24% High, 69% Very High). Fifty pts (91%) had a TP53 missense mutation in the DNA binding domain with multiple mutations in 18 (33%), and median variant allele frequency (VAF) of 25%. In 34 pts (62%), TP53 was the sole mutation. Median time on treatment is 154 days (11-392) with 8 pts ongoing. Eighteen pts (33%; 40% of evaluable pts) discontinued study treatment to proceed to allo-HSCT. Treatment (Tx)-related AEs in ≥ 20% of pts included nausea/vomiting (58%), dizziness (31%), constipation (24%), neuropathy (22%), leukopenia (22%) and thrombocytopenia (20%; all G1/G2 except cytopenias (G3/G4). Tx-related febrile neutropenia and anemia occurred in 9% and 5% of pts with no other G3/G4 event in >1 pt. Thirty and 60 day mortality was 2% (n=1) and 6% (n=3), respectively. At data cutoff, 45pts were response evaluable with a median follow up of 10.5 months (Fig 1A). ORR by IWG was 87% (39/45) with 24 CR (53%), 8 marrow CR (mCR)+HI (18%), 3 HI alone (7%), and 4 with mCR (9%). Of 6 non-responders, 4 had stable disease and 2 pts had progressive disease. Median time to response was 2.1 months (0.1-5.4) and median duration of response of 6.5 months. CR rate for MDS was 61% (20/33), 50% for AML (4/8) and 0% for MDS/MPN (0/4) with an 88% ORR rate for MDS/AML and 75% for MDS/MPN. An isolated mTP53 was predictive for a higher CR rate (69% vs 25%; P=.006) with a trend for higher ORR (93% vs 75%; P=.17). Additionally, pts with >10% p53 IHC+ BM-MNC was a covariate associated with higher CR rate (66% vs 13%; P=.01). Complete and partial cytogenetic response occurred in 41% (n=18) and 18% (n=8) of pts, respectively. On serial TP53 NGS using a VAF cutoff of 5%, 39% (n=21) of patients achieved NGS negativity, which was associated with improved OS (12.8 vs 9.2 months; P=.02). In NGS- pts, the median MRD VAF at maximum clearance was 0.63% (0.0%-5%) with 5 pts (11%) MRD negative. By intention-to-treat analysis, median OS was 11.6 months (95% CI 9.2-14) with significantly longer OS in responding pts (12.8 vs 3.9 months; P Conclusions: APR-246+AZA is a well-tolerated combination with high response rates in mTP53 MDS/AML. Response durations are promising accompanied by a high fraction of cytogenetic and deep molecular remissions leading to encouraging outcomes post-HSCT. These data support the ongoing, randomized phase 3 study of APR-246+AZA versus AZA alone in mTP53 MDS (NCT03745716). Disclosures Sallman: Abbvie: Speakers Bureau; Novartis: Speakers Bureau; Jazz: Research Funding; Incyte: Speakers Bureau; Celyad: Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding, Speakers Bureau. DeZern:Astex Pharmaceuticals, Inc.: Consultancy; Celgene: Consultancy. Garcia-Manero:Amphivena: Consultancy, Research Funding; Helsinn: Research Funding; Novartis: Research Funding; AbbVie: Research Funding; Celgene: Consultancy, Research Funding; Astex: Consultancy, Research Funding; Onconova: Research Funding; H3 Biomedicine: Research Funding; Merck: Research Funding. Steensma:Stemline: Consultancy; Pfizer: Consultancy; Aprea: Research Funding; H3 Biosciences: Other: Research funding to institution, not investigator.; Astex: Consultancy; Arrowhead: Equity Ownership; Onconova: Consultancy; Summer Road: Consultancy. Roboz:AbbVie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Actinium: Consultancy, Membership on an entity's Board of Directors or advisory committees; Agios: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amphivena: Consultancy, Membership on an entity's Board of Directors or advisory committees; Argenx: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astex: Consultancy, Membership on an entity's Board of Directors or advisory committees; Astellas: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bayer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celltrion: Consultancy, Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees; Eisai: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Jazz: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; MEI Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees; Orsenix: Consultancy, Membership on an entity's Board of Directors or advisory committees; Otsuka: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Roche/Genentech: Consultancy, Membership on an entity's Board of Directors or advisory committees; Sandoz: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees; Trovagene: Consultancy, Membership on an entity's Board of Directors or advisory committees. Sekeres:Syros: Membership on an entity's Board of Directors or advisory committees; Millenium: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees. Cluzeau:Jazz Pharma: Consultancy; Abbvie: Consultancy; Menarini: Consultancy. Sweet:Incyte: Research Funding; Celgene: Speakers Bureau; Pfizer: Consultancy; Agios: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Jazz: Speakers Bureau; Stemline: Consultancy; Abbvie: Membership on an entity's Board of Directors or advisory committees; Astellas: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Korbel:Aprea Therapeutics: Employment. Attar:Aprea Therapeutics: Employment. Kantarjian:Astex: Research Funding; Amgen: Honoraria, Research Funding; Ariad: Research Funding; BMS: Research Funding; AbbVie: Honoraria, Research Funding; Actinium: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Research Funding; Immunogen: Research Funding; Jazz Pharma: Research Funding; Takeda: Honoraria; Pfizer: Honoraria, Research Funding; Daiichi-Sankyo: Research Funding; Agios: Honoraria, Research Funding; Cyclacel: Research Funding. Lancet:Daiichi Sankyo: Consultancy, Other: fees for non-CME/CE services ; Agios, Biopath, Biosight, Boehringer Inglheim, Celator, Celgene, Janssen, Jazz Pharmaceuticals, Karyopharm, Novartis: Consultancy; Pfizer: Consultancy, Research Funding. Fenaux:Aprea: Research Funding; Astex: Honoraria, Research Funding; Celgene Corporation: Honoraria, Research Funding; Jazz: Honoraria, Research Funding. List:Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding. Komrokji:JAZZ: Consultancy; Novartis: Speakers Bureau; Incyte: Consultancy; DSI: Consultancy; pfizer: Consultancy; celgene: Consultancy; JAZZ: Speakers Bureau; Agios: Consultancy.
- Published
- 2019
- Full Text
- View/download PDF
39. Depletion of the Long Non-Coding RNA MALAT1 primes Chronic Myelomonocytic Leukemia (CMML) for Differentiation Therapy with All-Trans retinoic Acid (ATRA) through the Transcription Factor CREB
- Author
-
Eric Solary, Pearlie K. Epling-Burnette, Rami S. Komrokji, Nana Ben-Crentsil, Thomas Cluzeau, Jeffrey S. Painter, A. Robert MacLeod, Ariel Quintana, Hailing Zhang, Abhishek Dhawan, Maria E. Balasis, Hannah Newman, Alexis Vedder, Traci Kruer, Christopher Letson, Nathalie Droin, Jane Merlevede, Alan F. List, Eric Padron, Kathy L. McGraw, Sheng Wei, and Lei Sun
- Subjects
biology ,Immunology ,Retinoic acid ,Chronic myelomonocytic leukemia ,RNA ,Cell Biology ,Hematology ,medicine.disease ,CREB ,Biochemistry ,chemistry.chemical_compound ,Leukemia ,chemistry ,Tretinoin ,Differentiation therapy ,medicine ,Cancer research ,biology.protein ,Transcription factor ,medicine.drug - Abstract
Improvements in antisense technology have now enabled clinically relevant therapeutic credentialing of the noncoding genome. MALAT1 is a long non-coding RNA that, among other functions, is thought to serve as a nuclear scaffold for splicing and transcription factors. MALAT1 expression is associated with inferior prognosis across solid tumors and its depletion impairs proliferation and metastasis in preclinical solid tumor models. We found that elevated MALAT1 levels are independently associated with inferior overall survival in patients with CMML. Further, RNA-sequencing of primary CMML monocytes identified MALAT1 as the fourth most over-expressed transcript compared to controls. Therefore, we explored the biologic relevance and therapeutic candidacy of MALAT1 across several human and murine models of CMML. First, we crossed NRASQ61R/+Mx1-cre driven mice, which display a CMML-like phenotype, to MALAT1KO/KOmice. Although MALAT1KO/KOmice did not have abnormalities in complete blood counts, immunophenotyping of the hematopoietic stem cell (HSC) compartment identified statistically significantly lower numbers of HSC compared to wild type (WT) controls and a non-significant decrease in NRASQ61R/+/MALAT1KO/KOcompared to NRASQ61R/+alone. This decrease in HSC was not a result of impaired self-renewal as no differences were observed in these models after in vivo competitive transplant experiments. Therefore, we reasoned that MALAT1 expression may be controlling HSC differentiation. To test this, we transformed bone marrow cells from these models with an estrogen-regulated (ER) Hoxb8 construct enabling cells to maintain an HSC state until ER is withdrawn and myeloid differentiation is induced. ER-Hoxb8NRASQ61R/+/MALAT1KO/KOcells had increased basal levels of Gr-1 compared to ER-Hoxb8 transformed NRASQ61R/+alone that was dramatically enhanced upon ER withdrawal suggesting that MALAT1 depletion regulates myeloid differentiation. These findings were validated by assessment of morphology, transcriptome, and in vivo immunophenotyping of bone marrow and spleen cells. Further, moribund NRASQ61R/+/MALAT1KO/KOmice displayed a reduction in organomegaly typically associated with leukemic burden. We validated this in human monocytic leukemia by generating MALAT1 depleted THP-1 isogeneic cell lines using the CRISPR/Cas9 system. MALAT1 depleted THP-1 cells (MKO) had greater terminal differentiation according to immunophenotypic markers and morphology that was greatly enhanced when treated with phorbol myristate acetate. Last, MKO orthotopic xenografts demonstrated inferior human leukemia engraftment and decreased spleen and liver weights, and heterotopic xenografts exhibited reduced tumor volume, collectively suggesting diminished leukemic burden. Because ATRA has been clinically tested in CMML with minimal effects, we next explored whether MALAT1 depletion could potentiate ATRA differentiation in CMML. First, we treated MKO cells with ATRA and observed a large induction of myeloid differentiation by marker expression and morphologic assessment compared to isogenic controls. This was validated by NRASQ61R/+/MALAT1KO/KOmice demonstrating that ATRA more robustly induced differentiation compared to vehicle which was not seen in NRASQ61R/+/MALAT1+/+mice. Next, we tested MALAT1 antisense oligonucleotides (ASOs) currently under clinical development in THP-1 cells +/- ATRA and demonstrated both an increase in myeloid differentiation and apoptosis compared to ATRA alone. To test this therapeutic strategy in primary CMML specimens, we generated CMML patient-derived xenografts (n=30 mice) and treated each with ASO, ATRA, the combination, or controls and identified a more robust reduction in human HSC engraftment with the combination. To explore the mechanistic basis for these findings, we performed RNA-sequencing of MALAT1-depleted or control cells and identified that CREB target genes were differentially expressed. Basal protein levels of p-CREB were also decreased in MKO cells and were further reduced in the nucleus of MKO by western and microscopy. Lastly, overexpression of WT or constitutively active CREB but not its dominant negative rescued the differentiation effect seen in ATRA treated MKO cells. Taken together, MALAT1 is a novel, CREB-dependent regulator of myeloid differentiation and its depletion potentiates ATRA therapy. Disclosures Cluzeau: Menarini: Consultancy; Jazz Pharma: Consultancy; Abbvie: Consultancy. Komrokji:celgene: Consultancy; pfizer: Consultancy; DSI: Consultancy; JAZZ: Speakers Bureau; Novartis: Speakers Bureau; JAZZ: Consultancy; Agios: Consultancy; Incyte: Consultancy. MacLeod:Ionis Pharmaceuticals: Employment. List:Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding. Epling-Burnette:Forma Therapeutics: Research Funding; Celgene Corporation: Patents & Royalties, Research Funding; Incyte Corporation: Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
40. Combined Treatment with Lenalidomide and Epoetin Alfa Leads to Durable Responses in Patients with Epo-Refractory, Lower Risk Non-Deletion 5q [Del(5q)] MDS: Final Results of the E2905 Intergroup Phase III Study - an ECOG-ACRIN Cancer Research Group Study, Grant CA180820, and the National Cancer Institute of the National Institutes of Health
- Author
-
Rami S. Komrokji, Selina M. Luger, Jessica K. Altman, John M. Bennett, Martin S. Tallman, Jaroslaw P. Maciejewski, Kathy L. McGraw, Mark R. Litzow, David F. Claxton, Puneet Cheema, Charles A. Schiffer, Ryan J. Mattison, Andrew S. Artz, Amit Verma, Zhuoxin Sun, Alan F. List, and Timothy R. Wassenaar
- Subjects
medicine.medical_specialty ,Intention-to-treat analysis ,Group study ,business.industry ,Immunology ,Epoetin alfa ,Cell Biology ,Hematology ,Lower risk ,Biochemistry ,Combined treatment ,Family medicine ,medicine ,Clinical endpoint ,In patient ,business ,health care economics and organizations ,medicine.drug ,Lenalidomide - Abstract
Background: Only a small subset of Lower risk (LR) MDS patients benefit from treatment with rhu-Erythropoietin (Epo). We previously reported that lenalidomide (LEN) restores sensitivity to Epo in MDS progenitors by inducing the formation of lipid rafts that are enriched for signaling competent, JAK2/Epo-receptor complexes (McGraw K, et. al. PLoS One 2014; Basiorka A, et. al. Cancer Res 2016). In the MDS-002 and MDS-005 trials, treatment with LEN monotherapy gave rise to RBC transfusion-independence (TI) in 26% of azanucleoside-naïve, transfusion-dependent (TD) LR, non-del(5q) MDS patients for a median of 10.2 and 7.75 months, respectively. In a pilot study of Epo-refractory LR-MDS patients, the addition of epoetin alfa (EA) to LEN treatment yielded erythroid responses in 28% of patients who were unresponsive to LEN alone, suggesting that LEN may overcome clinical resistance to augment response to rhEpo (Komrokji R, et. al. Blood 2012). To test this hypothesis, we performed a randomized phase III trial comparing treatment with LEN to LEN+EA in LR non-del(5q) MDS patients who were refractory to, or not candidates for treatment with rhEpo. Methods: Patients with Low or Intermediate-1 IPSS risk MDS with hemoglobin 2 units/month) with serum Epo >500mU/mL were eligible. Patients were stratified by serum Epo level and prior rhEpo (EA vs. darbepoetin vs. none) then randomized to treatment with LEN 10 mg/d x21d q4wk (Arm A) or LEN + EA 60,000U SC/wk (Arm B). The primary endpoint was major erythroid response (MER) at week 16 which was defined according to transfusion status at baseline: (1) achievement of RBC-TI for ≥ 8 consecutive weeks AND a sustained ≥1 g/dL hemoglobin rise compared to mean pre-transfusion baseline value in TD patients; and (2) a >2 g/dL rise in hemoglobin without transfusion for ≥ 8 consecutive weeks in non-TD patients ( Results: Among 205 patients randomized, 14 were excluded from the primary analysis due to a 4 month interruption in drug supply. Among the 195 evaluable patients, 96 were assigned to Arm A and 99 to Arm B. Baseline characteristics were well balanced between arms with 85% of patients heavily TD, receiving a median of 4 units/8 weeks. Overall, 93% of patients received prior treatment with Epo and 18% azanucleosides. In an intent to treat analysis, 28/99 patients (28.3%) in Arm B achieved MER compared to 11/96 (11.5%) in Arm A (P=0.004). Among 136 patients who completed 16 weeks of study treatment, 28/72 (38.9%) and 10/64 (15.6%) achieved MER, respectively (P=0.004). Forty-four Arm A non-responders crossed over to combination-therapy with 11 patients (25%) experiencing a MER. Multivariable logistic regression analysis identified only treatment with LEN + EA as an independent covariate for erythroid response (P=0.01). Responses were durable with MER median duration of 23.8 months in Arm B compared to 13 months in Arm A. There was no significant difference in the frequency or distribution of >Grade 3, non-hematologic adverse events between treatment arms. Two patients progressed to AML while on study (Arm A), and no thromboembolic events were reported. Conclusions: LEN restores sensitivity to rhEpo in otherwise refractory, LR-non-del(5q) MDS patients to yield a significantly higher frequency of durable major erythroid responses compared to LEN alone. The addition of LEN to EA treatment is an effective strategy for the management of Epo-refractory patients with a potential duration of benefit extending to years. Disclosures List: Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding. Verma:Janssen: Research Funding; BMS: Research Funding; Stelexis: Equity Ownership, Honoraria; Acceleron: Honoraria; Celgene: Honoraria. Maciejewski:Novartis: Consultancy; Alexion: Consultancy. Komrokji:JAZZ: Speakers Bureau; Novartis: Speakers Bureau; JAZZ: Consultancy; Agios: Consultancy; Incyte: Consultancy; DSI: Consultancy; celgene: Consultancy; pfizer: Consultancy. Luger:Onconova: Research Funding; Pfizer: Honoraria; Seattle Genetics: Research Funding; Cyslacel: Research Funding; Biosight: Research Funding; Ariad: Research Funding; Agios: Honoraria; Genetech: Research Funding; Jazz: Honoraria; Daichi Sankyo: Honoraria; Kura: Research Funding; Celgene: Research Funding. Mattison:Pfizer: Membership on an entity's Board of Directors or advisory committees. Altman:Abbvie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Theradex: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Glycomimetics: Consultancy, Honoraria, Other: Data Safety and Monitoring Committee; Agios: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; France Foundation: Speakers Bureau; Daiichi Sankyo: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; prIME Oncology: Speakers Bureau; PeerView: Speakers Bureau; Cancer Expert Now: Consultancy; Novartis: Consultancy; Biosight: Other: US Lead. Claxton:Astellas Pharma: Other: Pharma support of clinical studies; Merck Sharp & Dohme Corp.: Other: Pharma support of clinical studies; Cyclacel Pharmaceuticals, Inc.: Other: Pharma support of clinical studies; Medimmune Inc.: Other: Pharma support of clinical studies; Novartis Pharmaceuticals: Other: Pharma support of clinical studies; Celgene Corporation: Other: Pharma support of clinical studies; Incyte Corporation: Other: Cyclacel Pharmaceuticals, Inc; Daiichi Sankyo Co. and Ambit Biosciences Corp: Other: Pharma support of clinical studies. Artz:Miltenyi: Research Funding. Tallman:Daiichi-Sankyo: Consultancy, Membership on an entity's Board of Directors or advisory committees; ADC Therapeutics: Research Funding; Oncolyze: Consultancy, Membership on an entity's Board of Directors or advisory committees; Tetraphase: Consultancy, Membership on an entity's Board of Directors or advisory committees; UpToDate: Patents & Royalties; Jazz Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Cellerant: Research Funding; Rigel: Consultancy, Membership on an entity's Board of Directors or advisory committees; KAHR: Consultancy, Membership on an entity's Board of Directors or advisory committees; Biosight: Research Funding; Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Nohla: Consultancy, Membership on an entity's Board of Directors or advisory committees; Orsenix: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Delta Fly Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees; BioLineRx: Consultancy, Membership on an entity's Board of Directors or advisory committees. OffLabel Disclosure: Lenalidomide used for treatment non-del 5q myelodysplastic syndromes.
- Published
- 2019
- Full Text
- View/download PDF
41. STAT3 mutations indicate the presence of subclinical T-cell clones in a subset of aplastic anemia and myelodysplastic syndrome patients
- Author
-
Austin G. Kulasekararaj, Satu Mustjoki, Bartlomiej P Przychodzen, Hanna Rajala, Catherine Nissen, Sonja Lagström, Inés Gómez-Seguí, Dan Zhang, Thomas P. Loughran, Holleh D Husseinzadeh, Andres Jerez, Thomas L. Olson, Aleksandra Wodnar-Filipowicz, Manuel G. Afable, Alan F. List, Michael J. Clemente, Pekka Ellonen, Ghulam J. Mufti, Naoko Hosono, Francis R LeBlanc, Alan E. Lichtin, Hideki Makishima, Kathy L. McGraw, Jaroslaw P. Maciejewski, and André Tichelli
- Subjects
Adult ,Male ,STAT3 Transcription Factor ,medicine.medical_treatment ,Immunology ,Chronic myelomonocytic leukemia ,Context (language use) ,Cell Separation ,Kaplan-Meier Estimate ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Aplastic anemia ,Proportional Hazards Models ,030304 developmental biology ,Chromosome 7 (human) ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,Myelodysplastic syndromes ,Bone marrow failure ,Anemia, Aplastic ,Myeloid leukemia ,Immunosuppression ,Cell Biology ,Hematology ,Middle Aged ,Flow Cytometry ,medicine.disease ,3. Good health ,Leukemia, Large Granular Lymphocytic ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female - Abstract
Large granular lymphocyte leukemia (LGL) is often associated with immune cytopenias and can cooccur in the context of aplastic anemia (AA) and myelodysplastic syndromes (MDS). We took advantage of the recent description of signal transducer and activator of transcription 3 (STAT3) mutations in LGL clonal expansions to test, using sensitive methods, for the presence of these mutations in a large cohort of 367 MDS and 140 AA cases. STAT3 clones can be found not only in known LGL concomitant cases, but in a small proportion of unsuspected ones (7% AA and 2.5% MDS). In STAT3-mutated AA patients, an interesting trend toward better responses of immunosuppressive therapy and an association with the presence of human leukocyte antigen-DR15 were found. MDSs harboring a STAT3 mutant clone showed a lower degree of bone marrow cellularity and a higher frequency of developing chromosome 7 abnormalities. STAT3-mutant LGL clones may facilitate a persistently dysregulated autoimmune activation, responsible for the primary induction of bone marrow failure in a subset of AA and MDS patients.
- Published
- 2013
- Full Text
- View/download PDF
42. The role of p53 in myelodysplastic syndromes and acute myeloid leukemia: molecular aspects and clinical implications
- Author
-
Kathy L. McGraw, Ling Zhang, Alan F. List, and David A. Sallman
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Biology ,Gene mutation ,medicine.disease_cause ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Molecular Targeted Therapy ,neoplasms ,Genetic Association Studies ,Lenalidomide ,Mutation ,Myelodysplastic syndromes ,Cytogenetics ,Myeloid leukemia ,Hematology ,medicine.disease ,Prognosis ,Gene Expression Regulation, Neoplastic ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Myelodysplastic Syndromes ,Cancer research ,Erythropoiesis ,Tumor Suppressor Protein p53 ,medicine.drug ,Signal Transduction - Abstract
TP53 gene mutations occurring in patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are associated with high-risk karyotypes including 17p abnormalities, monosomal and complex cytogenetics. TP53 mutations in these disorders portend rapid disease progression and resistance to conventional therapeutics. Notably, the size of the TP53 mutant clone as measured by mutation allele burden is directly linked to overall survival (OS) confirming the importance of p53 as a negative prognostic variable. In nucleolar stress-induced ribosomopathies, such as del(5q) MDS, disassociation of MDM2 and p53 results in p53 accumulation in erythroid precursors manifested as erythroid hypoplasia. P53 antagonism by lenalidomide or other therapeutics such as antisense oligonucleotides, repopulates erythroid precursors and enhances effective erythropoiesis. These findings demonstrate that p53 is an intriguing therapeutic target that is currently under investigation in MDS and AML. This study reviews molecular advances in understanding the role of p53 in MDS and AML, and explores potential therapeutic strategies in this era of personalized medicine.
- Published
- 2016
43. TP53 suppression promotes erythropoiesis in del(5q) MDS, suggesting a targeted therapeutic strategy in lenalidomide-resistant patients
- Author
-
F. Joseph Daugherty, Ji-Hyun Lee, Rami S. Komrokji, Alan F. List, Ling Zhang, Najla Al Ali, Gisela Caceres, William J. Fulp, Sheng Wei, Richard A. Wells, Neil Littleton, Kathy L. McGraw, Joseph O. Johnson, Jacqueline Boultwood, Andrea Pellagatti, Lubomir Sokol, Larry J. Smith, Kenian Liu, Ashley A. Basiorka, Lan Min Zhang, and Bon Ham Yip
- Subjects
Molecular Sequence Data ,Drug Resistance ,Oligonucleotides ,Fluorescent Antibody Technique ,Biology ,Real-Time Polymerase Chain Reaction ,Dexamethasone ,Statistics, Nonparametric ,hemic and lymphatic diseases ,medicine ,Humans ,Erythropoiesis ,Lenalidomide ,In Situ Hybridization, Fluorescence ,Erythroid Precursor Cells ,Multidisciplinary ,Base Sequence ,Cell growth ,Myelodysplastic syndromes ,Sequence Analysis, DNA ,Biological Sciences ,medicine.disease ,Flow Cytometry ,Immunohistochemistry ,Thalidomide ,medicine.anatomical_structure ,Treatment Outcome ,Apoptosis ,Myelodysplastic Syndromes ,Cancer research ,Bone marrow ,Tumor Suppressor Protein p53 ,Glucocorticoid ,medicine.drug - Abstract
Stabilization of p53 in erythroid precursors in response to nucleosomal stress underlies the hypoplastic anemia in myelodysplastic syndromes (MDS) with chromosome 5q deletion [del(5q)]. We investigated whether cenersen, a clinically active 20-mer antisense oligonucleotide complementary to TP53 exon10, could suppress p53 expression and restore erythropoiesis in del(5q) MDS. Cenersen treatment of ribosomal protein S-14-deficient erythroblasts significantly reduced cellular p53 and p53-up-regulated modulator of apoptosis expression compared with controls, accompanied by a significant reduction in apoptosis and increased cell proliferation. In a two-stage erythroid differentiation assay, cenersen significantly suppressed nuclear p53 in bone marrow CD34+ cells isolated from patients with del(5q) MDS, whereas erythroid burst recovery increased proportionally to the magnitude of p53 suppression without evidence of del(5q) clonal suppression (r = −0.6; P = 0.005). To explore the effect of p53 suppression on erythropoiesis in vivo, dexamethasone, a glucocorticoid receptor-dependent p53 antagonist, was added to lenalidomide treatment in eight lower-risk, transfusion-dependent, del(5q) MDS patients with acquired drug resistance. Transfusion independence was restored in five patients accompanied by expansion of erythroid precursors and decreased cellular p53 expression. We conclude that targeted suppression of p53 could support effective erythropoiesis in lenalidomide-resistant del(5q) MDS.
- Published
- 2016
44. Loss of heterozygosity in 7q myeloid disorders: clinical associations and genomic pathogenesis
- Author
-
Anna M. Jankowska, Andrea Pellagatti, Jacqueline Boultwood, Alison R. Moliterno, Hideki Makishima, Azim M Mohamedali, Alan F. List, Austin G. Kulasekararaj, Jaroslaw P. Maciejewski, Andres Jerez, Bartlomiej P Przychodzen, Mikkael A. Sekeres, Yuka Sugimoto, Ramon V. Tiu, Amit Verma, Kathy L. McGraw, Seiji Kojima, Hideki Muramatsu, Valeria Visconte, Michael A. McDevitt, Ghulam J. Mufti, and Christine L. O'Keefe
- Subjects
Adult ,Male ,Immunology ,Loss of Heterozygosity ,Biology ,Biochemistry ,Somatic evolution in cancer ,Polymorphism, Single Nucleotide ,Loss of heterozygosity ,Cohort Studies ,medicine ,Humans ,Myeloid Cells ,Bone Marrow Diseases ,Genetic Association Studies ,Aged ,Chromosome 7 (human) ,Genetics ,Chromosome Aberrations ,Massive parallel sequencing ,Myeloid Neoplasia ,Genome, Human ,Myelodysplastic syndromes ,Hypocellular Myelodysplastic Syndrome ,Myeloid leukemia ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Uniparental disomy ,Myelodysplastic Syndromes ,Female ,Chromosome Deletion ,Chromosomes, Human, Pair 7 - Abstract
Loss of heterozygosity affecting chromosome 7q is common in acute myeloid leukemia and myelodysplastic syndromes, pointing toward the essential role of this region in disease phenotype and clonal evolution. The higher resolution offered by recently developed genomic platforms may be used to establish more precise clinical correlations and identify specific target genes. We analyzed a series of patients with myeloid disorders using recent genomic technologies (1458 by single-nucleotide polymorphism arrays [SNP-A], 226 by next-generation sequencing, and 183 by expression microarrays). Using SNP-A, we identified chromosome 7q loss of heterozygosity segments in 161 of 1458 patients (11%); 26% of chronic myelomonocytic leukemia patients harbored 7q uniparental disomy, of which 41% had a homozygous EZH2 mutation. In addition, we describe an SNP-A–isolated deletion 7 hypocellular myelodysplastic syndrome subset, with a high rate of progression. Using direct and parallel sequencing, we found no recurrent mutations in typically large deletion 7q and monosomy 7 patients. In contrast, we detected a markedly decreased expression of genes included in our SNP-A defined minimally deleted regions. Although a 2-hit model is present in most patients with 7q uniparental disomy and a myeloproliferative phenotype, haplodeficient expression of defined regions of 7q may underlie pathogenesis in patients with deletions and predominant dysplastic features.
- Published
- 2016
- Full Text
- View/download PDF
45. Naive T-cells in myelodysplastic syndrome display intrinsic human telomerase reverse transcriptase (hTERT) deficiency
- Author
-
Rami S. Komrokji, Sheng Wei, Xiubao Ren, Jaroslaw P. Maciejewski, Pearlie K. Epling-Burnette, Jeffrey S. Painter, Rangasudhagar Radhakrishnan, Dana E. Rollison, Thomas P. Loughran, Alan F. List, Adam W. Mailloux, Ronald Paquette, Lili Yang, Kathy L. McGraw, and Hideki Makishima
- Subjects
Adult ,Male ,Cancer Research ,Telomerase ,Myeloid ,Adolescent ,Cell division ,T-Lymphocytes ,Biology ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,hemic and lymphatic diseases ,Gene expression ,T lymphocyte ,medicine ,Humans ,Telomerase reverse transcriptase ,Aged ,Cell Proliferation ,030304 developmental biology ,Aged, 80 and over ,telomere ,0303 health sciences ,Hematology ,Middle Aged ,myelodysplastic syndrome ,Telomere ,Haematopoiesis ,medicine.anatomical_structure ,Bromodeoxyuridine ,Oncology ,Case-Control Studies ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Female ,bone marrow failure ,transcription - Abstract
Telomeres are specialized structures providing chromosome integrity during cellular division along with protection against premature senescence and apoptosis. Accelerated telomere attrition in patients with myelodysplastic syndrome (MDS) occurs by an undefined mechanism. Although the MDS clone originates within the myeloid compartment, T-lymphocytes display repertoire contraction and loss of naive T-cells. The replicative lifespan of T-cells is stringently regulated by telomerase activity. In MDS cases, we show that purified CD3+ T-cells have significantly shorter telomere length and reduced proliferative capacity upon stimulation compared with controls. To understand the mechanism, telomerase enzymatic activity and telomerase reverse transcriptase (hTERT), gene expression were compared in MDS cases (n=35) and healthy controls (n=42) within different T-cell compartments. Telomerase activity is greatest in naive T-cells illustrating the importance of telomere repair in homeostatic repertoire regulation. Compared with healthy controls, MDS cases had lower telomerase induction (P
- Published
- 2012
- Full Text
- View/download PDF
46. STAT3 mutations unify the pathogenesis of chronic lymphoproliferative disorders of NK cells and T-cell large granular lymphocyte leukemia
- Author
-
Kathy L. McGraw, Satu Mustjoki, Mikkael A. Sekeres, Hideki Makishima, Thomas L. Olson, Eric D. Hsi, Andres Jerez, Lisa Durkin, Bartlomiej P Przychodzen, Hanna Koskela, Kathryn M Guinta, Marcin W. Wlodarski, Manuel G. Afable, Michael J. Clemente, Kwok Peng Ng, Francis R LeBlanc, Dan Zhang, Thomas P. Loughran, Alan F. List, Jaroslaw P. Maciejewski, Kimmo Porkka, and Inés Gómez-Seguí
- Subjects
Adult ,Male ,STAT3 Transcription Factor ,Large granular lymphocytic leukemia ,Blotting, Western ,Immunology ,Lymphoproliferative disorders ,Apoptosis ,Enzyme-Linked Immunosorbent Assay ,Biology ,Gene mutation ,Real-Time Polymerase Chain Reaction ,Biochemistry ,Pathogenesis ,Young Adult ,Biomarkers, Tumor ,medicine ,Humans ,Cytotoxic T cell ,RNA, Messenger ,T-Cell Large Granular Lymphocyte Leukemia ,Aged ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Aged, 80 and over ,Lymphoid Neoplasia ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Cell Biology ,Hematology ,Middle Aged ,Flow Cytometry ,medicine.disease ,Lymphoproliferative Disorders ,Killer Cells, Natural ,Leukemia, Large Granular Lymphocytic ,Gene expression profiling ,Leukemia ,Mutation ,Female ,T-Lymphocytes, Cytotoxic - Abstract
Chronic lymphoproliferative disorders of natural killer cells (CLPD-NKs) and T-cell large granular lymphocytic leukemias (T-LGLs) are clonal lymphoproliferations arising from either natural killer cells or cytotoxic T lymphocytes (CTLs). We have investigated for distribution and functional significance of mutations in 50 CLPD-NKs and 120 T-LGL patients by direct sequencing, allele-specific PCR, and microarray analysis. STAT3 gene mutations are present in both T and NK diseases: approximately one-third of patients with each type of disorder convey these mutations. Mutations were found in exons 21 and 20, encoding the Src homology 2 domain. Patients with mutations are characterized by symptomatic disease (75%), history of multiple treatments, and a specific pattern of STAT3 activation and gene deregulation, including increased expression of genes activated by STAT3. Many of these features are also found in patients with wild-type STAT3, indicating that other mechanisms of STAT3 activation can be operative in these chronic lymphoproliferative disorders. Treatment with STAT3 inhibitors, both in wild-type and mutant cases, resulted in accelerated apoptosis. STAT3 mutations are frequent in large granular lymphocytes suggesting a similar molecular dysregulation in malignant chronic expansions of NK and CTL origin. STAT3 mutations may distinguish truly malignant lymphoproliferations involving T and NK cells from reactive expansions.
- Published
- 2012
- Full Text
- View/download PDF
47. Identification of a risk dependent microRNA expression signature in myelodysplastic syndromes
- Author
-
Kathy L. McGraw, Justine Clark, Hans Alder, Gerard J. Nuovo, Celia Sigua, Lubomir Sokol, Stefano Volinia, Lynn C. Moscinski, Chang Gong Liu, Alan F. List, Carlo M. Croce, Dung Tsa Chen, and Gisela Caceres
- Subjects
Oncology ,medicine.medical_specialty ,Myeloid ,Myelodysplastic syndromes ,Hematology ,MicroRNA Expression Profile ,Biology ,medicine.disease ,Lower risk ,Gene expression profiling ,medicine.anatomical_structure ,International Prognostic Scoring System ,hemic and lymphatic diseases ,Internal medicine ,Immunology ,medicine ,Risk factor ,Survival analysis - Abstract
The myelodysplastic syndromes (MDS) display both haematological and biological heterogeneity with variable leukaemia potential. MicroRNAs play an important role in tumour suppression and the regulation of self-renewal and differentiation of haematopoietic progenitors. Using a microarray platform, we evaluated microRNA expression from 44 patients with MDS and 17 normal controls. We identified a thirteen microRNA signature with statistically significant differential expression between normal and MDS specimens (P < 0·01), including down-regulation of members of the leukaemia-associated MIRLET7 family. A unique signature consisting of 10 microRNAs was closely associated with International Prognostic Scoring System (IPSS) risk category permitting discrimination between lower (Low/Intermediate-1) and higher risk (Intermediate-2/High) disease (P < 0·01). Selective overexpression of MIR181 family members was detected in higher risk MDS, indicating pathogenetic overlap with acute myeloid leukaemia. Survival analysis of an independent cohort of 22 IPSS lower risk MDS patients revealed a median survival of 3·5 years in patients with high expression of MIR181 family compared to 9·3 years in patients with low MIR181 expression (P = 0·002). Our pilot study suggested that analysis of microRNA expression profile offers diagnostic utility, and provide pathogenetic and prognostic discrimination in MDS.
- Published
- 2011
- Full Text
- View/download PDF
48. Genomic-DNA Exposed By Somatic Gene Mutations Engages the cGAS/STING Axis to License the NLRP3 Inflammasome in Myelodysplastic Syndromes
- Author
-
Benjamin S. Meyer, Nicole D. Vincelette, Ling Zhang, Matthew A. Rodrigues, Alan F. List, Amy F McLemore, Sheng Wei, Kathy L. McGraw, Erika A. Eksioglu, Hsin-An Hou, Alexis H Onimus, Neelkamal Chaudhary, and Grace A Ward
- Subjects
0301 basic medicine ,Interferon-stimulated gene ,Immunology ,Wild type ,Caspase 1 ,Inflammasome ,Cell Biology ,Hematology ,Gene mutation ,Biology ,Biochemistry ,Molecular biology ,03 medical and health sciences ,030104 developmental biology ,medicine ,IRF7 ,IRF3 ,Interferon type I ,medicine.drug - Abstract
Background: The pathogenesis of Myelodysplastic Syndromes (MDS) is linked to constitutive innate immune stimulation that converges upon the NLRP3 inflammasome to induce pyroptosis, a caspase-1 dependent cell death. We have shown that inflammasome assembly is initiated by both cell-extrinsic stimuli such as S100A9 elaborated by Myeloid-Derived Suppressor Cells (MDSC), as well as cell-intrinsic somatic gene mutations (SGM) (Basiorka A, et. al. Blood 2016). SGM of varied classes evoke replication stress caused by transcriptional pauses that can expose genomic DNA to cytosolic sensors through unresolved R-loops or micronuclei formation. The cGMP-AMP Synthase-Stimulator of Interferon Genes (cGAS-STING) is a cell-intrinsic DNA surveillance pathway recognizing both cytosolic pathogenic and autologous DNA, leading to interferon stimulated gene (ISG) transcription and NLRP3 inflammasome activation, key biological features of MDS (Pellagatti A, et. al. Blood 2006; 108:337.). Here, we investigate the contribution of genomic cytosolic DNA engagement by cGAS-STING to NLRP3 inflammasome activation in MDS. Methods: MDS patient and healthy donor bone marrow mononuclear cells (BMMC) were isolated by Ficoll®-Hipaque method from consented participants at the Moffitt Cancer Center or the National Taiwan University Hospital (NTUH). Immortalized murine C57BL/6 Tet2-/- and MX1Cre/SRSF2P95H as well as respective wild type (WT) control BMMCs were used as MDS SGM models. Results: We first assessed cGAS-STING activation in MDS BMMC by measuring ISG response by microarray, demonstrating profoundly increased expression of ISG15, CXCL10, Samd9l, and Ifi27l2 in MDS BMMC (n=213) compared to healthy control BMMC (n=20) (p Conclusion: These data indicate that cGAS-STING engages redundant sources of cytosolic genomic-DNA in MDS to initiate a Type I interferon response and NLRP3 inflammasome activation. Inhibition of the cGAS-STING axis may represent a novel therapeutic strategy for investigation in MDS. Disclosures List: Celgene: Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
49. Phase 1b/2 Combination Study of APR-246 and Azacitidine (AZA) in Patients with TP53 mutant Myelodysplastic Syndromes (MDS) and Acute Myeloid Leukemia (AML)
- Author
-
Alan F. List, Jeffrey E. Lancet, David P. Steensma, Roger Tell, Rami S. Komrokji, Amy E. DeZern, Thomas Cluzeau, Ling Zhang, Chirag K Bhagat, Mikkael A. Sekeres, Pierre Fenaux, David A. Sallman, Najla Al Ali, John Puskas, Kendra Sweet, Kathy L. McGraw, Gail J. Roboz, Guillermo Garcia-Manero, Amy F McLemore, Eric Padron, and Jiqiang Yao
- Subjects
medicine.medical_specialty ,Therapy related ,business.industry ,Myelodysplastic syndromes ,Immunology ,Azacitidine ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Fixed dose ,Cytogenetic Response ,Decreased appetite ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,In patient ,Disease assessment ,business ,030215 immunology ,medicine.drug - Abstract
Introduction: TP53 mutant (mTP53) MDS and AML, accounting for 5-10% of de novo MDS and 25-30% of therapy related MDS (t-MDS), represent a distinct molecular cohort with inferior outcomes. Hypomethylating agents (HMA) are preferred treatments for patients (pts) with these mutations, although with CR rates of only 20-30% and median OS of 6-12 months. APR-246 is a novel, first-in-class small molecule that selectively induces apoptosis in mTP53 cancer cells through mutant p53 protein re-activation by restoring the wild-type conformation, with single agent activity in mTP53 AML. We report the planned, completed Phase 1b results of APR-246+ azacitidine (AZA) in mTP53 MDS/AML. Methods: This is a multicenter Phase 1b/2 trial of APR-246+AZA in HMA naïve mTP53 MDS and oligoblastic AML (≤ 30% blasts) pts ≥ 18 years of age. Pts received APR-246 in a 3+3 dose escalation design (50, 75, 100 mg/kg lean body weight (equivalent to 4500mg fixed dose based on PK studies)) IV daily over 4 days in a lead-in phase (days -14 to -10) followed by the same dose of APR-246 (days 1-4) + AZA 75 mg/m2 SC/IV over 7 days (days 4-10 or 4-5 and 8-12) in 28 day cycles. The primary objective was to define safety and the recommended Phase 2 dose (RP2D), with AEs graded by CTCAE v4.03 and DLT assessment over 6 weeks. Secondary objectives included response by IWG 2006 criteria as well as serial next generation sequencing (NGS) and p53 IHC for evaluation of clonal suppression and remission depth as predictors of outcomes. For minimal residual disease (MRD) analysis, a custom target-capture NGS assay was developed using unique molecular Identifiers for error correction with a limit of detection of 0.1% with results validated by pt specific digital droplet PCR (ddPCR). Nanostring nCounter RNA expression analysis was conducted on a panel of 770 genes after the lead-in phase to assess transcriptional effects induced by APR-246. Results: As of July 30, 2018, 12 pts (42% male; median age 66 years (39-73)) were enrolled. Three pts had AML-MRC and 9 had MDS; all pts had poor risk cytogenetics (17% poor, 83% very poor) and higher risk disease by IPSS-R (25% high, 75% very high). T-MDS occurred in 5 pts (42%) and 7 pts (58%) were transfusion-dependent at baseline. Median BM blasts were 9% (4-30). Eleven of 12 pts (92%) had a TP53 missense mutation in the DNA binding domain with multiple mutations in 4/12 pts (33%). For 9/12 pts (75%), TP53 was the sole mutation. Median time on study is 176 days (41-298) with 7 pts ongoing. Treatment (Tx) related AEs during the lead-in phase (all G1) included nausea (n=5), neuropathy (n=5), decreased appetite (n=2), and dizziness (n=2) which were all transient. Tx related AEs occurring in > 1 pt in the combination phase included nausea/vomiting (n=6), dizziness (n=3), headache (n=3), neuropathy (n=3), fall (n=2), pruritus (n=2), thrombocytopenia (n=6), neutropenia (n=5), and leukopenia (n=4); all G1/G2 except cytopenias (G3/G4). No DLTs have occurred to date. Eleven of twelve pts were response evaluable with 1 pt discontinuing tx prior to 1st disease assessment (Fig 1A). ORR by IWG was 100% (11/11) with 9 CR (82%) and 2 marrow CR (mCR; 18%). Median time to first response was 70 days (4-91) and one CR patient achieved mCR and partial cytogenetic response after APR-246 lead-in prior to combination therapy. All CR pts had high p53 positivity by IHC at baseline (25-80%) which normalized on serial assessment with the 2mCR pts having Conclusions: APR-246+AZA combination is well tolerated in mTP53 MDS/AML. Responses have been achieved in all evaluable pts (82% CR) accompanied by deep molecular and durable remissions. The RP2D of APR-246 is a fixed dose of 4500mg days 1-4 in combination with AZA and phase 2 accrual has begun. Disclosures Sallman: Celgene: Research Funding, Speakers Bureau. Sweet:Agios: Consultancy; Phizer: Consultancy; Agios: Consultancy; BMS: Honoraria; Celgene: Honoraria, Speakers Bureau; Jazz: Speakers Bureau; Celgene: Honoraria, Speakers Bureau; BMS: Honoraria; Phizer: Consultancy; Novartis: Consultancy, Honoraria, Speakers Bureau; Novartis: Consultancy, Honoraria, Speakers Bureau; Astellas: Consultancy; Jazz: Speakers Bureau; Astellas: Consultancy. Cluzeau:Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; AbbVie: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pfizer: Speakers Bureau; Sanofi: Speakers Bureau; Menarini: Consultancy; Amgen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Jazz Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Sekeres:Opsona: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Opsona: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees. Roboz:Orsenix: Consultancy; Cellectis: Research Funding; Eisai: Consultancy; Astex Pharmaceuticals: Consultancy; Argenx: Consultancy; Pfizer: Consultancy; Bayer: Consultancy; Eisai: Consultancy; Sandoz: Consultancy; Jazz Pharmaceuticals: Consultancy; Jazz Pharmaceuticals: Consultancy; Novartis: Consultancy; Pfizer: Consultancy; Otsuka: Consultancy; Bayer: Consultancy; Aphivena Therapeutics: Consultancy; Celltrion: Consultancy; Argenx: Consultancy; Celgene Corporation: Consultancy; Daiichi Sankyo: Consultancy; Celltrion: Consultancy; Sandoz: Consultancy; Astex Pharmaceuticals: Consultancy; Aphivena Therapeutics: Consultancy; Orsenix: Consultancy; Otsuka: Consultancy; Roche/Genentech: Consultancy; Roche/Genentech: Consultancy; Janssen Pharmaceuticals: Consultancy; Daiichi Sankyo: Consultancy; Novartis: Consultancy; AbbVie: Consultancy; Janssen Pharmaceuticals: Consultancy; Celgene Corporation: Consultancy; AbbVie: Consultancy; Cellectis: Research Funding. Bhagat:Genoptix: Employment. Tell:Aprea Therapeutics: Employment. Fenaux:Celgene: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Jazz: Honoraria, Research Funding; Otsuka: Honoraria, Research Funding; Roche: Honoraria. List:Celgene: Research Funding. Komrokji:Novartis: Honoraria, Speakers Bureau; Celgene: Honoraria, Research Funding; Novartis: Honoraria, Speakers Bureau; Celgene: Honoraria, Research Funding; Novartis: Honoraria, Speakers Bureau; Novartis: Honoraria, Speakers Bureau.
- Published
- 2018
- Full Text
- View/download PDF
50. Oxidized Mitochondrial DNA Is a Catalyst and Biomarker of Pyroptotic Cell Death in Myelodysplastic Syndromes
- Author
-
Alexis H Onimus, Nicole D. Vincelette, Erika A. Eksioglu, Hsin-An Hou, Amy F McLemore, Farnoosh Abbas-Aghababazadeh, Kathy L. McGraw, Benjamin S. Meyer, Alan F. List, Alexandra Calescibetta, Grace A Ward, Thu Le Trinh, and Sheng Wei
- Subjects
Programmed cell death ,Mitochondrial DNA ,medicine.diagnostic_test ,Myelodysplastic syndromes ,Immunology ,Caspase 1 ,Pyroptosis ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Flow cytometry ,chemistry.chemical_compound ,Biomarker ,chemistry ,medicine ,Cancer research ,DNA - Abstract
Constitutive innate immune activation is a pathogenetic driver of Myelodysplastic Syndromes (MDS) that directs ineffective hematopoiesis by NLRP3 inflammasome (IFM) assembly and pyroptotic cell death. IFM activation involves recruitment of caspase-1 (casp1) through the adapter protein, ASC, to facilitate autocatalytic cleavage of the zymogen to its active form that is responsible for interleukin (IL)-1β maturation, membrane pore formation and pyroptosis. Oxidized mitochondrial DNA (ox-mtDNA) has been proposed to serve as an alarmin that can activate the IFM by interaction directly with NLRP3 or engagement by DNA sensors, Toll-like receptor 9 (TLR9) and Cyclic GMP-AMP synthase (cGAS). Upon cytolysis, ox-mtDNA is released, permitting interaction with pattern recognition receptors on neighboring cells (Grishman, Pediatric Research, 2012, Shimada, 2012, Immunity. Vollmer, 2004, Immunology). Here, we investigate ox-mtDNA as an IFM-activator and pyroptotic biomarker in MDS. Incubation of TLR9 expressing cell lines, SKM1 (high expresser) and U937 (moderate expresser) with 50ng/mL ox-mtDNA (ND1 gene, amplified with oxidized guanosine) induced IFM activation evidenced by increased p-NFkβ, casp1 and IL-1β cleavage, ASC oligomerization and liberation of ASC specks. Direct interaction of ox-mtDNA with NLRP3 was confirmed by NLRP3 immunoprecipitation followed by probing for mtDNA using ND1 and CYTB specific primers and GAPDH primers as negative genomic control; mtDNA oxidation status was confirmed by dot blot. Furthermore, significantly increased expression of interferon stimulated genes (ISG) was seen in MDS bone marrow (BM) specimens (p≤0.01) compared to normal donors indicating TLR9 and/or cGAS activation. Ox-mtDNA engagement of TLR9 and cGAS was confirmed in MDS specimens by IF colocalization with corresponding IFM activation, as well as in MDS somatic gene mutation murine models (Tet2, SRSF2, U2AF) vs. Wt controls. Evaluation of surface TLR9 by flow cytometry showed significantly increased membrane expression in MDS CD34+ BMMC (n=4) vs. healthy donors (n=13) (p Collectively, these data indicate that ox-mtDNA both directly engages NLRP3 and the DNA sensors TLR9/cGAS to induce IFM activation and pyroptosis, creating a feed forward inflammatory cascade that extends to neighboring cells. Ox-mtDNA may serve as a biomarker and companion diagnostic for pyroptosis execution in MDS. Disclosures List: Celgene: Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.