25 results on '"Jitkova Y"'
Search Results
2. Translational Control by 4E-BP1/2 Suppressor Proteins Regulates Mitochondrial Biosynthesis and Function during CD8 + T Cell Proliferation.
- Author
-
Dimitriou ID, Meiri D, Jitkova Y, Elford AR, Koritzinsky M, Schimmer AD, Ohashi PS, Sonenberg N, and Rottapel R
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Cell Cycle Proteins metabolism, Cell Proliferation, Mammals genetics, Mice, Organelle Biogenesis, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Protein Biosynthesis, Eukaryotic Initiation Factors genetics, Eukaryotic Initiation Factors metabolism, Phosphoproteins metabolism
- Abstract
CD8
+ T cell proliferation and differentiation into effector and memory states are high-energy processes associated with changes in cellular metabolism. CD28-mediated costimulation of T cells activates the PI3K/AKT/mammalian target of rapamycin signaling pathway and induces eukaryotic translation initiation factor 4E-dependent translation through the derepression by 4E-BP1 and 4E-BP2. In this study, we demonstrate that 4E-BP1/2 proteins are required for optimum proliferation of mouse CD8+ T cells and the development of an antiviral effector function. We show that translation of genes encoding mitochondrial biogenesis is impaired in T cells derived from 4E-BP1/2-deficient mice. Our findings demonstrate an unanticipated role for 4E-BPs in regulating a metabolic program that is required for cell growth and biosynthesis during the early stages of CD8+ T cell expansion., (Copyright © 2022 by The American Association of Immunologists, Inc.)- Published
- 2022
- Full Text
- View/download PDF
3. Correction: Activation of RAS/MAPK pathway confers MCL-1 mediated acquired resistance to BCL-2 inhibitor venetoclax in acute myeloid leukemia.
- Author
-
Zhang Q, Riley-Gillis B, Han L, Jia Y, Lodi A, Zhang H, Ganesan S, Pan R, Konoplev SN, Sweeney SR, Ryan JA, Jitkova Y, Dunner K Jr, Grosskurth SE, Vijay P, Ghosh S, Lu C, Ma W, Kurtz S, Ruvolo VR, Ma H, Weng CC, Ramage CL, Baran N, Shi C, Cai T, Davis RE, Battula VL, Mi Y, Wang J, DiNardo CD, Andreeff M, Tyner JW, Schimmer A, Letai A, Padua RA, Bueso-Ramos CE, Tiziani S, Leverson J, Popovic R, and Konopleva M
- Published
- 2022
- Full Text
- View/download PDF
4. Activation of RAS/MAPK pathway confers MCL-1 mediated acquired resistance to BCL-2 inhibitor venetoclax in acute myeloid leukemia.
- Author
-
Zhang Q, Riley-Gillis B, Han L, Jia Y, Lodi A, Zhang H, Ganesan S, Pan R, Konoplev SN, Sweeney SR, Ryan JA, Jitkova Y, Dunner K Jr, Grosskurth SE, Vijay P, Ghosh S, Lu C, Ma W, Kurtz S, Ruvolo VR, Ma H, Weng CC, Ramage CL, Baran N, Shi C, Cai T, Davis RE, Battula VL, Mi Y, Wang J, DiNardo CD, Andreeff M, Tyner JW, Schimmer A, Letai A, Padua RA, Bueso-Ramos CE, Tiziani S, Leverson J, Popovic R, and Konopleva M
- Subjects
- Cell Line, Tumor, Drug Resistance, Neoplasm, Humans, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, MAP Kinase Signaling System drug effects, Myeloid Cell Leukemia Sequence 1 Protein genetics, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Sulfonamides pharmacology, ras Proteins
- Abstract
Despite high initial response rates, acute myeloid leukemia (AML) treated with the BCL-2-selective inhibitor venetoclax (VEN) alone or in combinations commonly acquires resistance. We performed gene/protein expression, metabolomic and methylation analyses of isogenic AML cell lines sensitive or resistant to VEN, and identified the activation of RAS/MAPK pathway, leading to increased stability and higher levels of MCL-1 protein, as a major acquired mechanism of VEN resistance. MCL-1 sustained survival and maintained mitochondrial respiration in VEN-RE cells, which had impaired electron transport chain (ETC) complex II activity, and MCL-1 silencing or pharmacologic inhibition restored VEN sensitivity. In support of the importance of RAS/MAPK activation, we found by single-cell DNA sequencing rapid clonal selection of RAS-mutated clones in AML patients treated with VEN-containing regimens. In summary, these findings establish RAS/MAPK/MCL-1 and mitochondrial fitness as key survival mechanisms of VEN-RE AML and provide the rationale for combinatorial strategies effectively targeting these pathways., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
5. Venetoclax enhances T cell-mediated antileukemic activity by increasing ROS production.
- Author
-
Lee JB, Khan DH, Hurren R, Xu M, Na Y, Kang H, Mirali S, Wang X, Gronda M, Jitkova Y, MacLean N, Arruda A, Alaniz Z, Konopleva MY, Andreeff M, Minden MD, Zhang L, and Schimmer AD
- Subjects
- Adult, Antineoplastic Agents therapeutic use, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Cells, Cultured, Humans, Immunity, Cellular drug effects, Leukemia, Myeloid, Acute immunology, Reactive Oxygen Species immunology, Sulfonamides therapeutic use, T-Lymphocytes immunology, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Leukemia, Myeloid, Acute drug therapy, Sulfonamides pharmacology, T-Lymphocytes drug effects
- Abstract
Venetoclax, a Bcl-2 inhibitor, in combination with the hypomethylating agent azacytidine, achieves complete remission with or without count recovery in ∼70% of treatment-naive elderly patients unfit for conventional intensive chemotherapy. However, the mechanism of action of this drug combination is not fully understood. We discovered that venetoclax directly activated T cells to increase their cytotoxicity against acute myeloid leukemia (AML) in vitro and in vivo. Venetoclax enhanced T-cell effector function by increasing reactive oxygen species generation through inhibition of respiratory chain supercomplexes formation. In addition, azacytidine induced a viral mimicry response in AML cells by activating the STING/cGAS pathway, thereby rendering the AML cells more susceptible to T cell-mediated cytotoxicity. Similar findings were seen in patients treated with venetoclax, as this treatment increased reactive oxygen species generation and activated T cells. Collectively, this study presents a new immune-mediated mechanism of action for venetoclax and azacytidine in the treatment of AML and highlights a potential combination of venetoclax and adoptive cell therapy for patients with AML., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
6. Mitochondrial carrier homolog 2 is necessary for AML survival.
- Author
-
Khan DH, Mullokandov M, Wu Y, Voisin V, Gronda M, Hurren R, Wang X, MacLean N, Jeyaraju DV, Jitkova Y, Xu GW, Laister R, Seneviratne A, Blatman ZM, Ketela T, Bader GD, Marhon SA, De Carvalho DD, Minden MD, Gross A, and Schimmer AD
- Subjects
- Acetylation, Animals, CRISPR-Cas Systems, Cell Differentiation, Cell Line, Tumor, Cell Nucleus metabolism, Fetal Blood cytology, Gene Expression Regulation, Leukemic genetics, Gene Knockdown Techniques, Histones metabolism, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Mice, Mice, Inbred C57BL, Myeloid-Lymphoid Leukemia Protein physiology, Oncogene Proteins, Fusion physiology, Protein Processing, Post-Translational, Pyruvic Acid metabolism, RNA Interference, RNA, Small Interfering genetics, RNA, Small Interfering pharmacology, Leukemia, Myeloid, Acute metabolism, Mitochondria metabolism, Mitochondrial Membrane Transport Proteins physiology, Neoplasm Proteins physiology
- Abstract
Through a clustered regularly insterspaced short palindromic repeats (CRISPR) screen to identify mitochondrial genes necessary for the growth of acute myeloid leukemia (AML) cells, we identified the mitochondrial outer membrane protein mitochondrial carrier homolog 2 (MTCH2). In AML, knockdown of MTCH2 decreased growth, reduced engraftment potential of stem cells, and induced differentiation. Inhibiting MTCH2 in AML cells increased nuclear pyruvate and pyruvate dehydrogenase (PDH), which induced histone acetylation and subsequently promoted the differentiation of AML cells. Thus, we have defined a new mechanism by which mitochondria and metabolism regulate AML stem cells and gene expression., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
7. Disrupting Mitochondrial Copper Distribution Inhibits Leukemic Stem Cell Self-Renewal.
- Author
-
Singh RP, Jeyaraju DV, Voisin V, Hurren R, Xu C, Hawley JR, Barghout SH, Khan DH, Gronda M, Wang X, Jitkova Y, Sharon D, Liyanagae S, MacLean N, Seneviratene AK, Mirali S, Borenstein A, Thomas GE, Soriano J, Orouji E, Minden MD, Arruda A, Chan SM, Bader GD, Lupien M, and Schimmer AD
- Subjects
- Cell Differentiation, Copper, Humans, Neoplastic Stem Cells, Cell Self Renewal, Leukemia, Myeloid, Acute
- Abstract
Leukemic stem cells (LSCs) rely on oxidative metabolism and are differentially sensitive to targeting mitochondrial pathways, which spares normal hematopoietic cells. A subset of mitochondrial proteins is folded in the intermembrane space via the mitochondrial intermembrane assembly (MIA) pathway. We found increased mRNA expression of MIA pathway substrates in acute myeloid leukemia (AML) stem cells. Therefore, we evaluated the effects of inhibiting this pathway in AML. Genetic and chemical inhibition of ALR reduces AML growth and viability, disrupts LSC self-renewal, and induces their differentiation. ALR inhibition preferentially decreases its substrate COX17, a mitochondrial copper chaperone, and knockdown of COX17 phenocopies ALR loss. Inhibiting ALR and COX17 increases mitochondrial copper levels which in turn inhibit S-adenosylhomocysteine hydrolase (SAHH) and lower levels of S-adenosylmethionine (SAM), DNA methylation, and chromatin accessibility to lower LSC viability. These results provide insight into mechanisms through which mitochondrial copper controls epigenetic status and viability of LSCs., Competing Interests: Declaration of Interests A.D.S. has received honoraria or consulting fees from Novartis, Jazz, Otsuka, and Takeda Pharmaceuticals and research support from Medivir AB and Takeda. A.D.S. owns stock in Abbvie Pharmaceuticals and is named on a patent application for the use of DNT cells for the treatment of leukemia. D.V.J is currently an employee of Celgene/Bristol-Myers Squibb (BMS). M.D.M. has received consulting fees from Astellas, Abbvie, Celgene/BMS, and GlaxoSmithKline (GSK). S.M.C. has received honoraria from Celgene and Agios. S.M.C. has received research funding from Agios, Celgene, and Abbvie Pharmaceuticals., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
8. The mitochondrial peptidase, neurolysin, regulates respiratory chain supercomplex formation and is necessary for AML viability.
- Author
-
Mirali S, Botham A, Voisin V, Xu C, St-Germain J, Sharon D, Hoff FW, Qiu Y, Hurren R, Gronda M, Jitkova Y, Nachmias B, MacLean N, Wang X, Arruda A, Minden MD, Horton TM, Kornblau SM, Chan SM, Bader GD, Raught B, and Schimmer AD
- Subjects
- Electron Transport, Humans, Metalloendopeptidases, Mitochondria metabolism, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Peptide Hydrolases metabolism
- Abstract
Neurolysin (NLN) is a zinc metallopeptidase whose mitochondrial function is unclear. We found that NLN was overexpressed in almost half of patients with acute myeloid leukemia (AML), and inhibition of NLN was selectively cytotoxic to AML cells and stem cells while sparing normal hematopoietic cells. Mechanistically, NLN interacted with the mitochondrial respiratory chain. Genetic and chemical inhibition of NLN impaired oxidative metabolism and disrupted the formation of respiratory chain supercomplexes (RCS). Furthermore, NLN interacted with the known RCS regulator, LETM1, and inhibition of NLN disrupted LETM1 complex formation. RCS were increased in patients with AML and positively correlated with NLN expression. These findings demonstrate that inhibiting RCS formation selectively targets AML cells and stem cells and highlights the therapeutic potential of pharmacologically targeting NLN in AML., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
9. De Novo Design of Boron-Based Peptidomimetics as Potent Inhibitors of Human ClpP in the Presence of Human ClpX.
- Author
-
Tan J, Grouleff JJ, Jitkova Y, Diaz DB, Griffith EC, Shao W, Bogdanchikova AF, Poda G, Schimmer AD, Lee RE, and Yudin AK
- Subjects
- Boronic Acids chemical synthesis, Boronic Acids metabolism, Drug Design, Endopeptidase Clp metabolism, Enzyme Assays, Humans, Peptide Library, Peptidomimetics chemical synthesis, Peptidomimetics metabolism, Protein Binding, Staphylococcus aureus enzymology, Stereoisomerism, Boronic Acids chemistry, Endopeptidase Clp antagonists & inhibitors, Peptidomimetics chemistry
- Abstract
Boronic acids have attracted the attention of synthetic and medicinal chemists due to boron's ability to modulate enzyme function. Recently, we demonstrated that boron-containing amphoteric building blocks facilitate the discovery of bioactive aminoboronic acids. Herein, we have augmented this capability with a de novo library design and a virtual screening platform modified for covalent ligands. This technique has allowed us to rapidly design and identify a series of α-aminoboronic acids as the first inhibitors of human ClpXP, which is responsible for the degradation of misfolded proteins.
- Published
- 2019
- Full Text
- View/download PDF
10. The Mitochondrial Transacylase, Tafazzin, Regulates AML Stemness by Modulating Intracellular Levels of Phospholipids.
- Author
-
Seneviratne AK, Xu M, Aristizabal Henao JJ, Fajardo VA, Hao Z, Voisin V, Xu GW, Hurren R, Kim S, MacLean N, Wang X, Gronda M, Jeyaraju D, Jitkova Y, Ketela T, Mullokandov M, Sharon D, Thomas G, Chouinard-Watkins R, Hawley JR, Schafer C, Yau HL, Khuchua Z, Aman A, Al-Awar R, Gross A, Claypool SM, Bazinet RP, Lupien M, Chan S, De Carvalho DD, Minden MD, Bader GD, Stark KD, LeBlanc P, and Schimmer AD
- Published
- 2019
- Full Text
- View/download PDF
11. Mitochondrial ClpP-Mediated Proteolysis Induces Selective Cancer Cell Lethality.
- Author
-
Ishizawa J, Zarabi SF, Davis RE, Halgas O, Nii T, Jitkova Y, Zhao R, St-Germain J, Heese LE, Egan G, Ruvolo VR, Barghout SH, Nishida Y, Hurren R, Ma W, Gronda M, Link T, Wong K, Mabanglo M, Kojima K, Borthakur G, MacLean N, Ma MCJ, Leber AB, Minden MD, Houry W, Kantarjian H, Stogniew M, Raught B, Pai EF, Schimmer AD, and Andreeff M
- Subjects
- Animals, Cell Line, Tumor, Cell Survival drug effects, Crystallography, X-Ray, Drug Screening Assays, Antitumor, Endopeptidase Clp chemistry, Female, HCT116 Cells, HEK293 Cells, Heterocyclic Compounds, 4 or More Rings chemistry, Heterocyclic Compounds, 4 or More Rings pharmacology, Humans, Imidazoles, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Mice, Models, Molecular, Point Mutation, Protein Conformation drug effects, Proteolysis, Pyridines, Pyrimidines, Tumor Suppressor Protein p53 metabolism, Xenograft Model Antitumor Assays, Endopeptidase Clp genetics, Endopeptidase Clp metabolism, Heterocyclic Compounds, 4 or More Rings administration & dosage, Leukemia, Myeloid, Acute drug therapy, Mitochondria metabolism
- Abstract
The mitochondrial caseinolytic protease P (ClpP) plays a central role in mitochondrial protein quality control by degrading misfolded proteins. Using genetic and chemical approaches, we showed that hyperactivation of the protease selectively kills cancer cells, independently of p53 status, by selective degradation of its respiratory chain protein substrates and disrupts mitochondrial structure and function, while it does not affect non-malignant cells. We identified imipridones as potent activators of ClpP. Through biochemical studies and crystallography, we show that imipridones bind ClpP non-covalently and induce proteolysis by diverse structural changes. Imipridones are presently in clinical trials. Our findings suggest a general concept of inducing cancer cell lethality through activation of mitochondrial proteolysis., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
12. The thymidine dideoxynucleoside analog, alovudine, inhibits the mitochondrial DNA polymerase γ, impairs oxidative phosphorylation and promotes monocytic differentiation in acute myeloid leukemia.
- Author
-
Yehudai D, Liyanage SU, Hurren R, Rizoska B, Albertella M, Gronda M, Jeyaraju DV, Wang X, Barghout SH, MacLean N, Siriwardena TP, Jitkova Y, Targett-Adams P, and Schimmer AD
- Subjects
- Animals, Antiviral Agents pharmacology, Apoptosis, Cell Proliferation, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Male, Mice, Mice, Inbred NOD, Mice, SCID, Mitochondria drug effects, Mitochondria metabolism, Monocytes drug effects, Monocytes metabolism, Thymidine chemistry, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Cell Differentiation drug effects, DNA Polymerase gamma antagonists & inhibitors, Dideoxynucleosides pharmacology, Leukemia, Myeloid, Acute drug therapy, Mitochondria pathology, Monocytes pathology, Oxidative Phosphorylation drug effects
- Abstract
Mitochondrial DNA encodes 13 proteins that comprise components of the respiratory chain that maintain oxidative phosphorylation. The replication of mitochondrial DNA is performed by the sole mitochondrial DNA polymerase γ. As acute myeloid leukemia (AML) cells and stem cells have an increased reliance on oxidative phosphorylation, we sought to evaluate polymerase γ inhibitors in AML. The thymidine dideoxynucleoside analog, alovudine, is an inhibitor of polymerase γ. In AML cells, alovudine depleted mitochondrial DNA, reduced mitochondrial encoded proteins, decreased basal oxygen consumption, and decreased cell proliferation and viability. To evaluate the effects of polymerase γ inhibition with alovudine in vivo , mice were xenografted with OCI-AML2 cells and then treated with alovudine. Systemic administration of alovudine reduced leukemic growth without evidence of toxicity and decreased levels of mitochondrial DNA in the leukemic cells. We also showed that alovudine increased the monocytic differentiation of AML cells. Genetic knockdown and other chemical inhibitors of polymerase γ also promoted AML differentiation, but the effects on AML differentiation were independent of reductions in oxidative phosphorylation or respiratory chain proteins. Thus, we have identified a novel mechanism by which mitochondria regulate AML fate and differentiation independent of oxidative phosphorylation. Moreover, we highlight polymerase γ inhibitors, such as alovudine, as novel therapeutic agents for AML., (Copyright© 2019 Ferrata Storti Foundation.)
- Published
- 2019
- Full Text
- View/download PDF
13. The Mitochondrial Transacylase, Tafazzin, Regulates for AML Stemness by Modulating Intracellular Levels of Phospholipids.
- Author
-
Seneviratne AK, Xu M, Henao JJA, Fajardo VA, Hao Z, Voisin V, Xu GW, Hurren R, Kim S, MacLean N, Wang X, Gronda M, Jeyaraju D, Jitkova Y, Ketela T, Mullokandov M, Sharon D, Thomas G, Chouinard-Watkins R, Hawley JR, Schafer C, Yau HL, Khuchua Z, Aman A, Al-Awar R, Gross A, Claypool SM, Bazinet RP, Lupien M, Chan S, De Carvalho DD, Minden MD, Bader GD, Stark KD, LeBlanc P, and Schimmer AD
- Subjects
- Acyltransferases, Animals, Cell Line, Tumor, Doxorubicin pharmacology, Female, Humans, Leukemia, Myeloid, Acute pathology, Male, Mice, Mice, Inbred NOD, Mice, SCID, Mice, Transgenic, Signal Transduction drug effects, Toll-Like Receptors metabolism, Transcription Factors antagonists & inhibitors, Transcription Factors deficiency, Leukemia, Myeloid, Acute metabolism, Mitochondria enzymology, Phospholipids metabolism, Transcription Factors metabolism
- Abstract
Tafazzin (TAZ) is a mitochondrial transacylase that remodels the mitochondrial cardiolipin into its mature form. Through a CRISPR screen, we identified TAZ as necessary for the growth and viability of acute myeloid leukemia (AML) cells. Genetic inhibition of TAZ reduced stemness and increased differentiation of AML cells both in vitro and in vivo. In contrast, knockdown of TAZ did not impair normal hematopoiesis under basal conditions. Mechanistically, inhibition of TAZ decreased levels of cardiolipin but also altered global levels of intracellular phospholipids, including phosphatidylserine, which controlled AML stemness and differentiation by modulating toll-like receptor (TLR) signaling., (Crown Copyright © 2019. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
14. A Phase 1 study of intravenous infusions of tigecycline in patients with acute myeloid leukemia.
- Author
-
Reed GA, Schiller GJ, Kambhampati S, Tallman MS, Douer D, Minden MD, Yee KW, Gupta V, Brandwein J, Jitkova Y, Gronda M, Hurren R, Shamas-Din A, Schuh AC, and Schimmer AD
- Subjects
- Adult, Aged, Aged, 80 and over, Antimetabolites, Antineoplastic adverse effects, Antimetabolites, Antineoplastic pharmacokinetics, Drug Monitoring, Drug Resistance, Neoplasm, Female, Humans, Infusions, Intravenous, Leukemia, Myeloid, Acute diagnosis, Male, Maximum Tolerated Dose, Middle Aged, Minocycline administration & dosage, Minocycline adverse effects, Minocycline pharmacokinetics, Mitochondria drug effects, Mitochondria metabolism, Recurrence, Retreatment, Risk Assessment, Tigecycline, Treatment Outcome, Antimetabolites, Antineoplastic administration & dosage, Leukemia, Myeloid, Acute drug therapy, Minocycline analogs & derivatives
- Abstract
Acute myeloid leukemia (AML) cells meet the higher energy, metabolic, and signaling demands of the cell by increasing mitochondrial biogenesis and mitochondrial protein translation. Blocking mitochondrial protein synthesis through genetic and chemical approaches kills human AML cells at all stages of development in vitro and in vivo. Tigecycline is an antimicrobial that we found inhibits mitochondrial protein synthesis in AML cells. Therefore, we conducted a phase 1 dose-escalation study of tigecycline administered intravenously daily 5 of 7 days for 2 weeks to patients with AML. A total of 27 adult patients with relapsed and refractory AML were enrolled in this study with 42 cycles being administered over seven dose levels (50-350 mg/day). Two patients experienced DLTs related to tigecycline at the 350 mg/day level resulting in a maximal tolerated dose of tigecycline of 300 mg as a once daily infusion. Pharmacokinetic experiments showed that tigecycline had a markedly shorter half-life in these patients than reported for noncancer patients. No significant pharmacodynamic changes or clinical responses were observed. Thus, we have defined the safety of once daily tigecycline in patients with refractory AML. Future studies should focus on schedules of the drug that permit more sustained target inhibition., (© 2016 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
15. Carnitine transporter CT2 (SLC22A16) is over-expressed in acute myeloid leukemia (AML) and target knockdown reduces growth and viability of AML cells.
- Author
-
Wu Y, Hurren R, MacLean N, Gronda M, Jitkova Y, Sukhai MA, Minden MD, and Schimmer AD
- Subjects
- Antineoplastic Agents pharmacology, Cell Cycle Checkpoints drug effects, Cell Line, Tumor, Cell Survival drug effects, Drug Synergism, Gene Knockdown Techniques, Humans, Oxygen metabolism, Cell Proliferation drug effects, Leukemia, Myeloid, Acute metabolism, Organic Cation Transport Proteins genetics, Organic Cation Transport Proteins metabolism, RNA, Small Interfering pharmacology
- Abstract
AML (acute myeloid leukemia) cells have a unique reliance on mitochondrial metabolism and fatty acid oxidation (FAO). Thus, blocking FAO is a potential therapeutic strategy to target these malignant cells. In the current study, we assessed plasma membrane carnitine transporters as novel therapeutic targets for AML. We examined the expression of the known plasma membrane carnitine transporters, OCTN1, OCTN2, and CT2 in AML cell lines and primary AML samples and compared expression to normal hematopoietic cells. Of the three carnitine transporters, CT2 demonstrated the greatest differential expression between AML and normal cells. Using shRNA, we knocked down CT2 and demonstrated that target knockdown impaired the function of the transporter. In addition, knockdown of CT2 reduced the growth and viability of AML cells with high expression of CT2 (OCI-AML2 and HL60), but not low expression. CT2 knockdown reduced basal oxygen consumption without a concomitant increase in glycolysis. Thus, CT2 may be a novel target for a subset of AML.
- Published
- 2015
- Full Text
- View/download PDF
16. Inhibition of the Mitochondrial Protease ClpP as a Therapeutic Strategy for Human Acute Myeloid Leukemia.
- Author
-
Cole A, Wang Z, Coyaud E, Voisin V, Gronda M, Jitkova Y, Mattson R, Hurren R, Babovic S, Maclean N, Restall I, Wang X, Jeyaraju DV, Sukhai MA, Prabha S, Bashir S, Ramakrishnan A, Leung E, Qia YH, Zhang N, Combes KR, Ketela T, Lin F, Houry WA, Aman A, Al-Awar R, Zheng W, Wienholds E, Xu CJ, Dick J, Wang JC, Moffat J, Minden MD, Eaves CJ, Bader GD, Hao Z, Kornblau SM, Raught B, and Schimmer AD
- Subjects
- Animals, Endopeptidase Clp metabolism, Heterografts, Humans, Male, Mice, Mice, Knockout, Mice, SCID, RNA, Small Interfering genetics, Endopeptidase Clp antagonists & inhibitors, Enzyme Inhibitors pharmacology, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute enzymology
- Abstract
From an shRNA screen, we identified ClpP as a member of the mitochondrial proteome whose knockdown reduced the viability of K562 leukemic cells. Expression of this mitochondrial protease that has structural similarity to the cytoplasmic proteosome is increased in leukemic cells from approximately half of all patients with AML. Genetic or chemical inhibition of ClpP killed cells from both human AML cell lines and primary samples in which the cells showed elevated ClpP expression but did not affect their normal counterparts. Importantly, Clpp knockout mice were viable with normal hematopoiesis. Mechanistically, we found that ClpP interacts with mitochondrial respiratory chain proteins and metabolic enzymes, and knockdown of ClpP in leukemic cells inhibited oxidative phosphorylation and mitochondrial metabolism., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
17. AML cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress.
- Author
-
Sriskanthadevan S, Jeyaraju DV, Chung TE, Prabha S, Xu W, Skrtic M, Jhas B, Hurren R, Gronda M, Wang X, Jitkova Y, Sukhai MA, Lin FH, Maclean N, Laister R, Goard CA, Mullen PJ, Xie S, Penn LZ, Rogers IM, Dick JE, Minden MD, and Schimmer AD
- Subjects
- Cell Death, Cell Respiration, Electron Transport, Humans, Mitochondrial Size, Reactive Oxygen Species metabolism, Tumor Cells, Cultured, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Oxidative Stress physiology, Oxygen Consumption physiology
- Abstract
Mitochondrial respiration is a crucial component of cellular metabolism that can become dysregulated in cancer. Compared with normal hematopoietic cells, acute myeloid leukemia (AML) cells and patient samples have higher mitochondrial mass, without a concomitant increase in respiratory chain complex activity. Hence these cells have a lower spare reserve capacity in the respiratory chain and are more susceptible to oxidative stress. We therefore tested the effects of increasing the electron flux through the respiratory chain as a strategy to induce oxidative stress and cell death preferentially in AML cells. Treatment with the fatty acid palmitate induced oxidative stress and cell death in AML cells, and it suppressed tumor burden in leukemic cell lines and primary patient sample xenografts in the absence of overt toxicity to normal cells and organs. These data highlight a unique metabolic vulnerability in AML, and identify a new therapeutic strategy that targets abnormal oxidative metabolism in this malignancy., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
18. A novel formulation of tigecycline has enhanced stability and sustained antibacterial and antileukemic activity.
- Author
-
Jitkova Y, Gronda M, Hurren R, Wang X, Goard CA, Jhas B, and Schimmer AD
- Subjects
- Animals, Anti-Bacterial Agents pharmacokinetics, Anti-Bacterial Agents pharmacology, Ascorbic Acid chemistry, Cell Line, Tumor, Cell Survival drug effects, Chromatography, High Pressure Liquid, Drug Stability, Humans, Immunoblotting, Mice, Mice, SCID, Minocycline chemistry, Minocycline pharmacokinetics, Minocycline pharmacology, Minocycline therapeutic use, Pyruvic Acid chemistry, Tigecycline, Anti-Bacterial Agents chemistry, Leukemia, Myeloid, Acute drug therapy, Minocycline analogs & derivatives
- Abstract
Tigecycline is a broad-spectrum, first-in-class glycylcycline antibiotic currently used to treat complicated skin and intra-abdominal infections, as well as community-acquired pneumonia. In addition, we have demonstrated that tigecycline also has in vitro and in vivo activity against acute myeloid leukemia (AML) due to its ability to inhibit mitochondrial translation. Tigecycline is relatively unstable after reconstitution, and this instability may limit the use of the drug in ambulatory infusions for the treatment of infection and may prevent the development of optimal dosing schedules for the treatment of AML. This study sought to identify a formulation that improved the stability of the drug after reconstitution and maintained its antimicrobial and antileukemic activity. A panel of chemical additives was tested to identify excipients that enhanced the stability of tigecycline in solution at room temperature for up to one week. We identified a novel formulation containing the oxygen-reducing agents ascorbic acid (3 mg/mL) and pyruvate (60 mg/mL), in saline solution, pH 7.0, in which tigecycline (1 mg/mL) remained intact when protected from light for at least 7 days. This formulation also preserved the drug's antibacterial and antileukemic activity in vitro. Moreover, the novel formulation retained tigecycline's antileukemic activity in vivo. Thus, we identified and characterized a novel formulation for tigecycline that preserves its stability and efficacy after reconstitution.
- Published
- 2014
- Full Text
- View/download PDF
19. Mapping the cellular response to small molecules using chemogenomic fitness signatures.
- Author
-
Lee AY, St Onge RP, Proctor MJ, Wallace IM, Nile AH, Spagnuolo PA, Jitkova Y, Gronda M, Wu Y, Kim MK, Cheung-Ong K, Torres NP, Spear ED, Han MK, Schlecht U, Suresh S, Duby G, Heisler LE, Surendra A, Fung E, Urbanus ML, Gebbia M, Lissina E, Miranda M, Chiang JH, Aparicio AM, Zeghouf M, Davis RW, Cherfils J, Boutry M, Kaiser CA, Cummins CL, Trimble WS, Brown GW, Schimmer AD, Bankaitis VA, Nislow C, Bader GD, and Giaever G
- Subjects
- Cell Line, Tumor, Haploinsufficiency, Humans, Pharmacogenetics, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae genetics, Cells drug effects, Drug Evaluation, Preclinical methods, Drug Resistance genetics, Gene Regulatory Networks, Genome-Wide Association Study methods, Small Molecule Libraries pharmacology
- Abstract
Genome-wide characterization of the in vivo cellular response to perturbation is fundamental to understanding how cells survive stress. Identifying the proteins and pathways perturbed by small molecules affects biology and medicine by revealing the mechanisms of drug action. We used a yeast chemogenomics platform that quantifies the requirement for each gene for resistance to a compound in vivo to profile 3250 small molecules in a systematic and unbiased manner. We identified 317 compounds that specifically perturb the function of 121 genes and characterized the mechanism of specific compounds. Global analysis revealed that the cellular response to small molecules is limited and described by a network of 45 major chemogenomic signatures. Our results provide a resource for the discovery of functional interactions among genes, chemicals, and biological processes.
- Published
- 2014
- Full Text
- View/download PDF
20. Oral ciclopirox olamine displays biological activity in a phase I study in patients with advanced hematologic malignancies.
- Author
-
Minden MD, Hogge DE, Weir SJ, Kasper J, Webster DA, Patton L, Jitkova Y, Hurren R, Gronda M, Goard CA, Rajewski LG, Haslam JL, Heppert KE, Schorno K, Chang H, Brandwein JM, Gupta V, Schuh AC, Trudel S, Yee KW, Reed GA, and Schimmer AD
- Subjects
- Administration, Oral, Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Antineoplastic Agents blood, Antineoplastic Agents pharmacokinetics, Ciclopirox, Female, Gastrointestinal Diseases chemically induced, Gene Expression Regulation, Neoplastic drug effects, Half-Life, Hematologic Neoplasms blood, Hematologic Neoplasms pathology, Humans, Inactivation, Metabolic, Inhibitor of Apoptosis Proteins genetics, Iron Chelating Agents administration & dosage, Iron Chelating Agents adverse effects, Iron Chelating Agents metabolism, Iron Chelating Agents pharmacokinetics, Male, Middle Aged, Neoplasm Proteins genetics, Pyridones adverse effects, Pyridones blood, Pyridones pharmacokinetics, RNA, Messenger blood, RNA, Neoplasm blood, Survivin, Treatment Outcome, Antineoplastic Agents therapeutic use, Hematologic Neoplasms drug therapy, Iron Chelating Agents therapeutic use, Pyridones therapeutic use, Salvage Therapy
- Abstract
The antimycotic ciclopirox olamine is an intracellular iron chelator that has anticancer activity in vitro and in vivo. We developed an oral formulation of ciclopirox olamine and conducted the first-in-human phase I study of this drug in patients with relapsed or refractory hematologic malignancies (Trial registration ID: NCT00990587). Patients were treated with 5-80 mg/m² oral ciclopirox olamine once daily for five days in 21-day treatment cycles. Pharmacokinetic and pharmacodynamic companion studies were performed in a subset of patients. Following definition of the half-life of ciclopirox olamine, an additional cohort was enrolled and treated with 80 mg/m² ciclopirox olamine four times daily. Adverse events and clinical response were monitored throughout the trial. Twenty-three patients received study treatment. Ciclopirox was rapidly absorbed and cleared with a short half-life. Plasma concentrations of an inactive ciclopirox glucuronide metabolite were greater than those of ciclopirox. Repression of survivin expression was observed in peripheral blood cells isolated from patients treated once daily with ciclopirox olamine at doses greater than 10 mg/m², demonstrating biological activity of the drug. Dose-limiting gastrointestinal toxicities were observed in patients receiving 80 mg/m² four times daily, and no dose limiting toxicity was observed at 40 mg/m² once daily. Hematologic improvement was observed in two patients. Once-daily dosing of oral ciclopirox olamine was well tolerated in patients with relapsed or refractory hematologic malignancies, and further optimization of dosing regimens is warranted in this patient population., (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
21. A porphodimethene chemical inhibitor of uroporphyrinogen decarboxylase.
- Author
-
Yip KW, Zhang Z, Sakemura-Nakatsugawa N, Huang JW, Vu NM, Chiang YK, Lin CL, Kwan JY, Yue S, Jitkova Y, To T, Zahedi P, Pai EF, Schimmer AD, Lovell JF, Sessler JL, and Liu FF
- Subjects
- Cell Line, Tumor, Cell Survival drug effects, Epithelial Cells drug effects, Humans, Inhibitory Concentration 50, Molecular Structure, Porphyrins chemistry, Porphyrins pharmacology, Recombinant Proteins chemistry, Substrate Specificity, Uroporphyrinogen Decarboxylase chemistry, Models, Molecular, Porphyrins chemical synthesis, Recombinant Proteins metabolism, Uroporphyrinogen Decarboxylase antagonists & inhibitors
- Abstract
Uroporphyrinogen decarboxylase (UROD) catalyzes the conversion of uroporphyrinogen to coproporphyrinogen during heme biosynthesis. This enzyme was recently identified as a potential anticancer target; its inhibition leads to an increase in reactive oxygen species, likely mediated by the Fenton reaction, thereby decreasing cancer cell viability and working in cooperation with radiation and/or cisplatin. Because there is no known chemical UROD inhibitor suitable for use in translational studies, we aimed to design, synthesize, and characterize such a compound. Initial in silico-based design and docking analyses identified a potential porphyrin analogue that was subsequently synthesized. This species, a porphodimethene (named PI-16), was found to inhibit UROD in an enzymatic assay (IC50 = 9.9 µM), but did not affect porphobilinogen deaminase (at 62.5 µM), thereby exhibiting specificity. In cellular assays, PI-16 reduced the viability of FaDu and ME-180 cancer cells with half maximal effective concentrations of 22.7 µM and 26.9 µM, respectively, and only minimally affected normal oral epithelial (NOE) cells. PI-16 also combined effectively with radiation and cisplatin, with potent synergy being observed in the case of cisplatin in FaDu cells (Chou-Talalay combination index <1). This work presents the first known synthetic UROD inhibitor, and sets the foundation for the design, synthesis, and characterization of higher affinity and more effective UROD inhibitors.
- Published
- 2014
- Full Text
- View/download PDF
22. Metabolic adaptation to chronic inhibition of mitochondrial protein synthesis in acute myeloid leukemia cells.
- Author
-
Jhas B, Sriskanthadevan S, Skrtic M, Sukhai MA, Voisin V, Jitkova Y, Gronda M, Hurren R, Laister RC, Bader GD, Minden MD, and Schimmer AD
- Subjects
- Anti-Bacterial Agents pharmacology, Cell Line, Tumor, Electron Transport Complex IV genetics, Gene Expression Regulation, Leukemic drug effects, Gene Expression Regulation, Leukemic genetics, Glycolysis drug effects, Glycolysis genetics, Humans, Hypoxia-Inducible Factor 1, alpha Subunit biosynthesis, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Minocycline analogs & derivatives, Minocycline pharmacology, Mitochondrial Proteins genetics, Neoplasm Proteins genetics, Oxygen Consumption drug effects, Oxygen Consumption genetics, Tigecycline, Drug Resistance, Neoplasm, Electron Transport Complex IV biosynthesis, Leukemia, Myeloid, Acute metabolism, Mitochondrial Proteins biosynthesis, Neoplasm Proteins biosynthesis, Protein Biosynthesis
- Abstract
Recently, we demonstrated that the anti-bacterial agent tigecycline preferentially induces death in leukemia cells through the inhibition of mitochondrial protein synthesis. Here, we sought to understand mechanisms of resistance to tigecycline by establishing a leukemia cell line resistant to the drug. TEX leukemia cells were treated with increasing concentrations of tigecycline over 4 months and a population of cells resistant to tigecycline (RTEX+TIG) was selected. Compared to wild type cells, RTEX+TIG cells had undetectable levels of mitochondrially translated proteins Cox-1 and Cox-2, reduced oxygen consumption and increased rates of glycolysis. Moreover, RTEX+TIG cells were more sensitive to inhibitors of glycolysis and more resistant to hypoxia. By electron microscopy, RTEX+TIG cells had abnormally swollen mitochondria with irregular cristae structures. RNA sequencing demonstrated a significant over-representation of genes with binding sites for the HIF1α:HIF1β transcription factor complex in their promoters. Upregulation of HIF1α mRNA and protein in RTEX+TIG cells was confirmed by Q-RTPCR and immunoblotting. Strikingly, upon removal of tigecycline from RTEX+TIG cells, the cells re-established aerobic metabolism. Levels of Cox-1 and Cox-2, oxygen consumption, glycolysis, mitochondrial mass and mitochondrial membrane potential returned to wild type levels, but HIF1α remained elevated. However, upon re-treatment with tigecycline for 72 hours, the glycolytic phenotype was re-established. Thus, we have generated cells with a reversible metabolic phenotype by chronic treatment with an inhibitor of mitochondrial protein synthesis. These cells will provide insight into cellular adaptations used to cope with metabolic stress.
- Published
- 2013
- Full Text
- View/download PDF
23. A phase I study of the metal ionophore clioquinol in patients with advanced hematologic malignancies.
- Author
-
Schimmer AD, Jitkova Y, Gronda M, Wang Z, Brandwein J, Chen C, Gupta V, Schuh A, Yee K, Chen J, Ackloo S, Booth T, Keays S, and Minden MD
- Subjects
- Adult, Aged, Aged, 80 and over, Clioquinol adverse effects, Clioquinol blood, Clioquinol pharmacokinetics, Drug Administration Schedule, Female, Follow-Up Studies, Hematologic Neoplasms blood, Hematologic Neoplasms metabolism, Humans, Ionophores adverse effects, Ionophores pharmacokinetics, Male, Middle Aged, Proteasome Endopeptidase Complex metabolism, Clioquinol administration & dosage, Hematologic Neoplasms drug therapy, Ionophores administration & dosage
- Abstract
Unlabelled: Clioquinol is a small-molecule metal ionophore that inhibits the proteasome through a metal-dependent mechanism. Here, we report a phase I study of clioquinol in patients with refractory hematologic malignancies. Neuropathy and abdominal pain were dose-limiting toxicities. Minimal pharmacodynamic effects were observed, and there were no clinical responses., Background: Clioquinol is a small-molecule metal ionophore that inhibits the enzymatic activity of the proteasome and displays preclinical efficacy in hematologic malignancies in vitro and in vivo. Therefore, we conducted a phase I clinical trial of clioquinol in patients with refractory hematologic malignancies to assess its safety and determine its biological activity in this patient population., Methods: Patients with refractory hematologic malignancies were treated with increasing doses of oral clioquinol twice daily for 15 doses. Plasma and intracellular levels of clioquinol were measured. Enzymatic activity of the proteasome was measured before and after drug administration., Results: Sixteen cycles of clioquinol were administered to 11 patients with 5 patients reenrolled at the next dose level as per the permitted intrapatient dose escalation. Dose-limiting neurotoxicity and abdominal pain were observed at a dose of 1600 mg twice daily. Intracellular drug levels were low. Minimal inhibition of the proteasome was observed. No clinical responses were observed., Conclusion: In patients with refractory hematologic malignancies, the maximal tolerated dose of clioquinol was determined. Minimal inhibition of the proteasome was observed at tolerable doses, likely due to low intracellular levels of the drug., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
24. Wnt inhibitor screen reveals iron dependence of β-catenin signaling in cancers.
- Author
-
Song S, Christova T, Perusini S, Alizadeh S, Bao RY, Miller BW, Hurren R, Jitkova Y, Gronda M, Isaac M, Joseph B, Subramaniam R, Aman A, Chau A, Hogge DE, Weir SJ, Kasper J, Schimmer AD, Al-awar R, Wrana JL, and Attisano L
- Subjects
- Acute Disease, Administration, Oral, Benzoates pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Ciclopirox, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Deferasirox, Deferoxamine pharmacology, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, HEK293 Cells, Humans, Hydrazones chemistry, Iron Chelating Agents pharmacology, Leukemia, Myeloid drug therapy, Leukemia, Myeloid genetics, Leukemia, Myeloid pathology, Oligonucleotide Array Sequence Analysis, Promoter Regions, Genetic genetics, Pyridones administration & dosage, Pyridones therapeutic use, Reverse Transcriptase Polymerase Chain Reaction, Triazoles pharmacology, Wnt Proteins genetics, Wnt Proteins metabolism, beta Catenin genetics, Hydrazones pharmacology, Iron metabolism, Wnt Proteins antagonists & inhibitors, Wnt Signaling Pathway drug effects, beta Catenin metabolism
- Abstract
Excessive signaling from the Wnt pathway is associated with numerous human cancers. Using a high throughput screen designed to detect inhibitors of Wnt/β-catenin signaling, we identified a series of acyl hydrazones that act downstream of the β-catenin destruction complex to inhibit both Wnt-induced and cancer-associated constitutive Wnt signaling via destabilization of β-catenin. We found that these acyl hydrazones bind iron in vitro and in intact cells and that chelating activity is required to abrogate Wnt signaling and block the growth of colorectal cancer cell lines with constitutive Wnt signaling. In addition, we found that multiple iron chelators, desferrioxamine, deferasirox, and ciclopirox olamine similarly blocked Wnt signaling and cell growth. Moreover, in patients with AML administered ciclopirox olamine, we observed decreased expression of the Wnt target gene AXIN2 in leukemic cells. The novel class of acyl hydrazones would thus be prime candidates for further development as chemotherapeutic agents. Taken together, our results reveal a critical requirement for iron in Wnt signaling and they show that iron chelation serves as an effective mechanism to inhibit Wnt signaling in humans.
- Published
- 2011
- Full Text
- View/download PDF
25. Inhibition of mitochondrial translation as a therapeutic strategy for human acute myeloid leukemia.
- Author
-
Skrtić M, Sriskanthadevan S, Jhas B, Gebbia M, Wang X, Wang Z, Hurren R, Jitkova Y, Gronda M, Maclean N, Lai CK, Eberhard Y, Bartoszko J, Spagnuolo P, Rutledge AC, Datti A, Ketela T, Moffat J, Robinson BH, Cameron JH, Wrana J, Eaves CJ, Minden MD, Wang JC, Dick JE, Humphries K, Nislow C, Giaever G, and Schimmer AD
- Subjects
- Animals, Cell Line, Tumor, Humans, Mice, Minocycline pharmacology, Mitochondrial Proteins genetics, Peptide Elongation Factor Tu genetics, RNA, Small Interfering, Saccharomyces cerevisiae drug effects, Tigecycline, Antineoplastic Agents pharmacology, Genes, Mitochondrial, Leukemia drug therapy, Minocycline analogs & derivatives, Mitochondria drug effects, Neoplastic Stem Cells drug effects, Protein Biosynthesis drug effects
- Abstract
To identify FDA-approved agents targeting leukemic cells, we performed a chemical screen on two human leukemic cell lines and identified the antimicrobial tigecycline. A genome-wide screen in yeast identified mitochondrial translation inhibition as the mechanism of tigecycline-mediated lethality. Tigecycline selectively killed leukemia stem and progenitor cells compared to their normal counterparts and also showed antileukemic activity in mouse models of human leukemia. ShRNA-mediated knockdown of EF-Tu mitochondrial translation factor in leukemic cells reproduced the antileukemia activity of tigecycline. These effects were derivative of mitochondrial biogenesis that, together with an increased basal oxygen consumption, proved to be enhanced in AML versus normal hematopoietic cells and were also important for their difference in tigecycline sensitivity., (2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.