1
|
Boila LD, Ghosh S, Bandyopadhyay SK, Jin L, Murison A, Zeng AGX, Shaikh W, Bhowmik S, Muddineni SSNA, Biswas M, Sinha S, Chatterjee SS, Mbong N, Gan OI, Bose A, Chakraborty S, Arruda A, Kennedy JA, Mitchell A, Lechman ER, Banerjee D, Milyavsky M, Minden MD, Dick JE, Sengupta A. KDM6 demethylases integrate DNA repair gene regulation and loss of KDM6A sensitizes human acute myeloid leukemia to PARP and BCL2 inhibition. Leukemia 2023; 37:751-764. [PMID: 36720973 DOI: 10.1038/s41375-023-01833-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous, aggressive malignancy with dismal prognosis and with limited availability of targeted therapies. Epigenetic deregulation contributes to AML pathogenesis. KDM6 proteins are histone-3-lysine-27-demethylases that play context-dependent roles in AML. We inform that KDM6-demethylase function critically regulates DNA-damage-repair-(DDR) gene expression in AML. Mechanistically, KDM6 expression is regulated by genotoxic stress, with deficiency of KDM6A-(UTX) and KDM6B-(JMJD3) impairing DDR transcriptional activation and compromising repair potential. Acquired KDM6A loss-of-function mutations are implicated in chemoresistance, although a significant percentage of relapsed-AML has upregulated KDM6A. Olaparib treatment reduced engraftment of KDM6A-mutant-AML-patient-derived xenografts, highlighting synthetic lethality using Poly-(ADP-ribose)-polymerase-(PARP)-inhibition. Crucially, a higher KDM6A expression is correlated with venetoclax tolerance. Loss of KDM6A increased mitochondrial activity, BCL2 expression, and sensitized AML cells to venetoclax. Additionally, BCL2A1 associates with venetoclax resistance, and KDM6A loss was accompanied with a downregulated BCL2A1. Corroborating these results, dual targeting of PARP and BCL2 was superior to PARP or BCL2 inhibitor monotherapy in inducing AML apoptosis, and primary AML cells carrying KDM6A-domain mutations were even more sensitive to the combination. Together, our study illustrates a mechanistic rationale in support of a novel combination therapy for AML based on subtype-heterogeneity, and establishes KDM6A as a molecular regulator for determining therapeutic efficacy.
Collapse
Affiliation(s)
- Liberalis Debraj Boila
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Subhadeep Ghosh
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, West Bengal, India
| | - Subham K Bandyopadhyay
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, West Bengal, India
| | - Liqing Jin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Andy G X Zeng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Wasim Shaikh
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, West Bengal, India
| | - Satyaki Bhowmik
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, West Bengal, India
| | | | - Mayukh Biswas
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Irving Cancer Research Center, Columbia University Medical Center, New York, NY, 10032, USA
| | - Sayantani Sinha
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Shankha Subhra Chatterjee
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Nathan Mbong
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Olga I Gan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Anwesha Bose
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India.,Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, West Bengal, India
| | - Sayan Chakraborty
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - James A Kennedy
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada.,Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Amanda Mitchell
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Debasis Banerjee
- Park Clinic, Gorky Terrace and Ramakrishna Mission Seva Pratisthan, Kolkata, 700017, West Bengal, India
| | - Michael Milyavsky
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada.,Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, M5G 2C4, Canada.,Department of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| | - Amitava Sengupta
- Stem Cell & Leukemia Lab, CSIR-Indian Institute of Chemical Biology, IICB-Translational Research Unit of Excellence, Salt Lake, Kolkata, 700091, West Bengal, India. .,Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, West Bengal, India. .,CSIR-IICB-Cancer Biology & Inflammatory Disorder Division, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, West Bengal, India.
| |
Collapse
|
2
|
Lee JC, Liu S, Wang Y, Liang Y, Jablons DM. MK256 is a novel CDK8 inhibitor with potent antitumor activity in AML through downregulation of the STAT pathway. Oncotarget 2022; 13:1217-1236. [PMCID: PMC9629815 DOI: 10.18632/oncotarget.28305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most lethal form of AML due to disease relapse. Cyclin dependent kinase 8 (CDK8) is a serine/threonine kinase that belongs to the family of Cyclin-dependent kinases and is an emerging target for the treatment of AML. MK256, a potent, selective, and orally available CDK8 inhibitor was developed to target AML. We sought to examine the anticancer effect of MK256 on AML. In CD34+/CD38- leukemia stem cells, we found that MK256 induced differentiation and maturation. Treatment of MK256 inhibited proliferation of AML cell lines. Further studies of the inhibitory effect suggested that MK256 not only downregulated phosphorylated STAT1(S727) and STAT5(S726), but also lowered mRNA expressions of MCL-1 and CCL2 in AML cell lines. Efficacy of MK256 was shown in MOLM-14 xenograft models, and the inhibitory effect on phosphorylated STAT1(S727) and STAT5(S726) with treatment of MK256 was observed in vivo. Pharmacologic dynamics study of MK256 in MOLM-14 xenograft models showed dose-dependent inhibition of the STAT pathway. Both in vitro and in vivo studies suggested that MK256 could effectively downregulate the STAT pathway. In vitro ADME, pharmacological kinetics, and toxicity of MK256 were profiled to evaluate the drug properties of MK256. Our results show that MK256 is a novel CDK8 inhibitor with a desirable efficacy and safety profile and has great potential to be a promising drug candidate for AML through regulating the STAT pathway.
Collapse
Affiliation(s)
- Jen-Chieh Lee
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA,2Touro University, College of Osteopathic Medicine, Vallejo, CA 94592, USA,*These authors contributed equally to this work,Correspondence to:Jen-Chieh Lee, email:
| | - Shu Liu
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA,*These authors contributed equally to this work,Shu Liu, email:
| | - Yucheng Wang
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA,*These authors contributed equally to this work
| | - You Liang
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - David M. Jablons
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
3
|
Thomas GE, Egan G, García-Prat L, Botham A, Voisin V, Patel PS, Hoff FW, Chin J, Nachmias B, Kaufmann KB, Khan DH, Hurren R, Wang X, Gronda M, MacLean N, O'Brien C, Singh RP, Jones CL, Harding SM, Raught B, Arruda A, Minden MD, Bader GD, Hakem R, Kornblau S, Dick JE, Schimmer AD. The metabolic enzyme hexokinase 2 localizes to the nucleus in AML and normal haematopoietic stem and progenitor cells to maintain stemness. Nat Cell Biol 2022; 24:872-884. [PMID: 35668135 PMCID: PMC9203277 DOI: 10.1038/s41556-022-00925-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 04/22/2022] [Indexed: 11/21/2022]
Abstract
Mitochondrial metabolites regulate leukaemic and normal stem cells by affecting epigenetic marks. How mitochondrial enzymes localize to the nucleus to control stem cell function is less understood. We discovered that the mitochondrial metabolic enzyme hexokinase 2 (HK2) localizes to the nucleus in leukaemic and normal haematopoietic stem cells. Overexpression of nuclear HK2 increases leukaemic stem cell properties and decreases differentiation, whereas selective nuclear HK2 knockdown promotes differentiation and decreases stem cell function. Nuclear HK2 localization is phosphorylation-dependent, requires active import and export, and regulates differentiation independently of its enzymatic activity. HK2 interacts with nuclear proteins regulating chromatin openness, increasing chromatin accessibilities at leukaemic stem cell-positive signature and DNA-repair sites. Nuclear HK2 overexpression decreases double-strand breaks and confers chemoresistance, which may contribute to the mechanism by which leukaemic stem cells resist DNA-damaging agents. Thus, we describe a non-canonical mechanism by which mitochondrial enzymes influence stem cell function independently of their metabolic function. Thomas, Egan et al. report that hexokinase 2 localizes to the nucleus of leukaemic and normal haematopoietic cells to maintain stemness by interacting with nuclear proteins and modulating chromatin accessibility independently of its kinase activity.
Collapse
Affiliation(s)
- Geethu Emily Thomas
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Grace Egan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Laura García-Prat
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Aaron Botham
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Veronique Voisin
- Terrence Donnelly Centre for Cellular and Biomedical Research, University of Toronto, Toronto, Ontario, Canada
| | - Parasvi S Patel
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Fieke W Hoff
- Department of Pediatric Hematology/Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Jordan Chin
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Boaz Nachmias
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Kerstin B Kaufmann
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Dilshad H Khan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Xiaoming Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Neil MacLean
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Cristiana O'Brien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Rashim P Singh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Courtney L Jones
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shane M Harding
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Gary D Bader
- Terrence Donnelly Centre for Cellular and Biomedical Research, University of Toronto, Toronto, Ontario, Canada
| | - Razq Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Steve Kornblau
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Nachmias B, Khan DH, Voisin V, Mer AS, Thomas GE, Segev N, St-Germain J, Hurren R, Gronda M, Botham A, Wang X, Maclean N, Seneviratne AK, Duong N, Xu C, Arruda A, Orouji E, Algouneh A, Hakem R, Shlush L, Minden MD, Raught B, Bader GD, Schimmer AD. IPO11 regulates the nuclear import of BZW1/2 and is necessary for AML cells and stem cells. Leukemia 2022; 36:1283-1295. [PMID: 35152270 PMCID: PMC9061300 DOI: 10.1038/s41375-022-01513-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 01/06/2022] [Accepted: 01/26/2022] [Indexed: 12/24/2022]
Abstract
AML cells are arranged in a hierarchy with stem/progenitor cells giving rise to more differentiated bulk cells. Despite the importance of stem/progenitors in the pathogenesis of AML, the determinants of the AML stem/progenitor state are not fully understood. Through a comparison of genes that are significant for growth and viability of AML cells by way of a CRISPR screen, with genes that are differentially expressed in leukemia stem cells (LSC), we identified importin 11 (IPO11) as a novel target in AML. Importin 11 (IPO11) is a member of the importin β family of proteins that mediate transport of proteins across the nuclear membrane. In AML, knockdown of IPO11 decreased growth, reduced engraftment potential of LSC, and induced differentiation. Mechanistically, we identified the transcription factors BZW1 and BZW2 as novel cargo of IPO11. We further show that BZW1/2 mediate a transcriptional signature that promotes stemness and survival of LSC. Thus, we demonstrate for the first time how specific cytoplasmic-nuclear regulation supports stem-like transcriptional signature in relapsed AML.
Collapse
|
5
|
Shukla S, Ying W, Gray F, Yao Y, Simes ML, Zhao Q, Miao H, Cho HJ, González-Alonso P, Winkler A, Lund G, Purohit T, Kim E, Zhang X, Ray JM, He S, Nikolaidis C, Ndoj J, Wang J, Jaremko Ł, Jaremko M, Ryan RJH, Guzman ML, Grembecka J, Cierpicki T. Small-molecule inhibitors targeting Polycomb repressive complex 1 RING domain. Nat Chem Biol 2021; 17:784-793. [PMID: 34155404 PMCID: PMC8238916 DOI: 10.1038/s41589-021-00815-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/10/2021] [Indexed: 12/11/2022]
Abstract
Polycomb repressive complex 1 (PRC1) is an essential chromatin-modifying complex that monoubiquitinates histone H2A and is involved in maintaining the repressed chromatin state. Emerging evidence suggests PRC1 activity in various cancers, rationalizing the need for small-molecule inhibitors with well-defined mechanisms of action. Here, we describe the development of compounds that directly bind to RING1B-BMI1, the heterodimeric complex constituting the E3 ligase activity of PRC1. These compounds block the association of RING1B-BMI1 with chromatin and inhibit H2A ubiquitination. Structural studies demonstrate that these inhibitors bind to RING1B by inducing the formation of a hydrophobic pocket in the RING domain. Our PRC1 inhibitor, RB-3, decreases the global level of H2A ubiquitination and induces differentiation in leukemia cell lines and primary acute myeloid leukemia (AML) samples. In summary, we demonstrate that targeting the PRC1 RING domain with small molecules is feasible, and RB-3 represents a valuable chemical tool to study PRC1 biology.
Collapse
Affiliation(s)
- Shirish Shukla
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Weijiang Ying
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Felicia Gray
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yiwu Yao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Miranda L Simes
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Qingjie Zhao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Hongzhi Miao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Hyo Je Cho
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Alyssa Winkler
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - George Lund
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Trupta Purohit
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - EunGi Kim
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xiaotian Zhang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Joshua M Ray
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Shihan He
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Juliano Ndoj
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jingya Wang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- MedImmune, LLC, Gaithersburg, MD, USA
| | - Łukasz Jaremko
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Mariusz Jaremko
- Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Russell J H Ryan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Monica L Guzman
- Division of Hematology and Medical Oncology, Leukemia Program, Weill Cornell Medicine/New York-Presbyterian Hospital, New York, NY, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
6
|
Very long chain fatty acid metabolism is required in acute myeloid leukemia. Blood 2021; 137:3518-3532. [PMID: 33720355 DOI: 10.1182/blood.2020008551] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/21/2021] [Indexed: 12/28/2022] Open
Abstract
Acute myeloid leukemia (AML) cells have an atypical metabolic phenotype characterized by increased mitochondrial mass, as well as a greater reliance on oxidative phosphorylation and fatty acid oxidation (FAO) for survival. To exploit this altered metabolism, we assessed publicly available databases to identify FAO enzyme overexpression. Very long chain acyl-CoA dehydrogenase (VLCAD; ACADVL) was found to be overexpressed and critical to leukemia cell mitochondrial metabolism. Genetic attenuation or pharmacological inhibition of VLCAD hindered mitochondrial respiration and FAO contribution to the tricarboxylic acid cycle, resulting in decreased viability, proliferation, clonogenic growth, and AML cell engraftment. Suppression of FAO at VLCAD triggered an increase in pyruvate dehydrogenase activity that was insufficient to increase glycolysis but resulted in adenosine triphosphate depletion and AML cell death, with no effect on normal hematopoietic cells. Together, these results demonstrate the importance of VLCAD in AML cell biology and highlight a novel metabolic vulnerability for this devastating disease.
Collapse
|
7
|
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, Schimmer AD. The mitochondrial peptidase, neurolysin, regulates respiratory chain supercomplex formation and is necessary for AML viability. Sci Transl Med 2021; 12:12/538/eaaz8264. [PMID: 32269163 DOI: 10.1126/scitranslmed.aaz8264] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 03/09/2020] [Indexed: 12/18/2022]
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.
Collapse
Affiliation(s)
- Sara Mirali
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada.,Institute of Medical Science, University of Toronto, Toronto M5S 1A8, Ontario, Canada
| | - Aaron Botham
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Ontario, Canada
| | - Veronique Voisin
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario M5S 3E1, Canada
| | - Changjiang Xu
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario M5S 3E1, Canada
| | | | - David Sharon
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Fieke W Hoff
- Department of Pediatric Oncology/Hematology, University Medical Center Groningen, Groningen 9700 RB, Netherlands.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yihua Qiu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rose Hurren
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Yulia Jitkova
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Boaz Nachmias
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Neil MacLean
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Xiaoming Wang
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Andrea Arruda
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada.,Institute of Medical Science, University of Toronto, Toronto M5S 1A8, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Ontario, Canada
| | - Terzah M Horton
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX 77030, USA
| | - Steven M Kornblau
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Steven M Chan
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Ontario, Canada
| | - Gary D Bader
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario M5S 3E1, Canada.,Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Ontario, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada. .,Institute of Medical Science, University of Toronto, Toronto M5S 1A8, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Ontario, Canada
| |
Collapse
|
8
|
Mitochondrial carrier homolog 2 is necessary for AML survival. Blood 2021; 136:81-92. [PMID: 32299104 DOI: 10.1182/blood.2019000106] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/18/2020] [Indexed: 01/04/2023] Open
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.
Collapse
|
9
|
Polak A, Bialopiotrowicz E, Krzymieniewska B, Wozniak J, Stojak M, Cybulska M, Kaniuga E, Mikula M, Jablonska E, Gorniak P, Noyszewska-Kania M, Szydlowski M, Piechna K, Piwocka K, Bugajski L, Lech-Maranda E, Barankiewicz J, Kolkowska-Lesniak A, Patkowska E, Glodkowska-Mrowka E, Baran N, Juszczynski P. SYK inhibition targets acute myeloid leukemia stem cells by blocking their oxidative metabolism. Cell Death Dis 2020; 11:956. [PMID: 33159047 PMCID: PMC7648638 DOI: 10.1038/s41419-020-03156-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022]
Abstract
Spleen tyrosine kinase (SYK) is an important oncogene and signaling mediator activated by cell surface receptors crucial for acute myeloid leukemia (AML) maintenance and progression. Genetic or pharmacologic inhibition of SYK in AML cells leads to increased differentiation, reduced proliferation, and cellular apoptosis. Herein, we addressed the consequences of SYK inhibition to leukemia stem-cell (LSC) function and assessed SYK-associated pathways in AML cell biology. Using gain-of-function MEK kinase mutant and constitutively active STAT5A, we demonstrate that R406, the active metabolite of a small-molecule SYK inhibitor fostamatinib, induces differentiation and blocks clonogenic potential of AML cells through the MEK/ERK1/2 pathway and STAT5A transcription factor, respectively. Pharmacological inhibition of SYK with R406 reduced LSC compartment defined as CD34+CD38−CD123+ and CD34+CD38−CD25+ in vitro, and decreased viability of LSCs identified by a low abundance of reactive oxygen species. Primary leukemic blasts treated ex vivo with R406 exhibited lower engraftment potential when xenotransplanted to immunodeficient NSG/J mice. Mechanistically, these effects are mediated by disturbed mitochondrial biogenesis and suppression of oxidative metabolism (OXPHOS) in LSCs. These mechanisms appear to be partially dependent on inhibition of STAT5 and its target gene MYC, a well-defined inducer of mitochondrial biogenesis. In addition, inhibition of SYK increases the sensitivity of LSCs to cytarabine (AraC), a standard of AML induction therapy. Taken together, our findings indicate that SYK fosters OXPHOS and participates in metabolic reprogramming of AML LSCs in a mechanism that at least partially involves STAT5, and that SYK inhibition targets LSCs in AML. Since active SYK is expressed in a majority of AML patients and confers inferior prognosis, the combination of SYK inhibitors with standard chemotherapeutics such as AraC constitutes a new therapeutic modality that should be evaluated in future clinical trials.
Collapse
Affiliation(s)
- Anna Polak
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Emilia Bialopiotrowicz
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Beata Krzymieniewska
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Jolanta Wozniak
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
| | - Magdalena Cybulska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Ewelina Kaniuga
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michał Mikula
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Ewa Jablonska
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Patryk Gorniak
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Monika Noyszewska-Kania
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Maciej Szydlowski
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Karolina Piechna
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Lukasz Bugajski
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Lech-Maranda
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Joanna Barankiewicz
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | | | - Elzbieta Patkowska
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Eliza Glodkowska-Mrowka
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Przemyslaw Juszczynski
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland.
| |
Collapse
|
10
|
Mirali S, Schimmer AD. The role of mitochondrial proteases in leukemic cells and leukemic stem cells. Stem Cells Transl Med 2020; 9:1481-1487. [PMID: 32761807 PMCID: PMC7695628 DOI: 10.1002/sctm.20-0142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/18/2020] [Accepted: 05/30/2020] [Indexed: 12/13/2022] Open
Abstract
The biological function of most mitochondrial proteases has not been well characterized. Moreover, most of the available information on the normal function of these proteases has been derived from studies in model organisms. Recently, the mitochondrial proteases caseinolytic protease P (CLPP) and neurolysin (NLN) have been identified as therapeutic targets in acute myeloid leukemia (AML). Both proteases are overexpressed in approximately 40% of AML patients. Mechanistically, CLPP and NLN maintain the integrity of the mitochondrial respiratory chain: CLPP cleaves defective respiratory chain proteins, while NLN promotes the formation of respiratory chain supercomplexes. In this review, we highlight the functional consequences of inhibiting and activating mitochondrial proteases and discuss their potential as therapeutic targets in AML.
Collapse
Affiliation(s)
- Sara Mirali
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
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, Schimmer AD. Disrupting Mitochondrial Copper Distribution Inhibits Leukemic Stem Cell Self-Renewal. Cell Stem Cell 2020; 26:926-937.e10. [PMID: 32416059 DOI: 10.1016/j.stem.2020.04.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 02/27/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022]
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.
Collapse
Affiliation(s)
- Rashim Pal Singh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Danny V Jeyaraju
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Changjiang Xu
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - James R Hawley
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Samir H Barghout
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Dilshad H Khan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xiaoming Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yulia Jitkova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David Sharon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sanduni Liyanagae
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Neil MacLean
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Sara Mirali
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Adina Borenstein
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Geethu E Thomas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Joelle Soriano
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Elias Orouji
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Steven M Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
12
|
van Galen P, Mbong N, Kreso A, Schoof EM, Wagenblast E, Ng SWK, Krivdova G, Jin L, Nakauchi H, Dick JE. Integrated Stress Response Activity Marks Stem Cells in Normal Hematopoiesis and Leukemia. Cell Rep 2019; 25:1109-1117.e5. [PMID: 30380403 DOI: 10.1016/j.celrep.2018.10.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/25/2018] [Accepted: 10/02/2018] [Indexed: 12/20/2022] Open
Abstract
Lifelong maintenance of the blood system requires equilibrium between clearance of damaged hematopoietic stem cells (HSCs) and long-term survival of the HSC pool. Severe perturbations of cellular homeostasis result in rapid HSC loss to maintain clonal purity. However, normal homeostatic processes can also generate lower-level stress; how HSCs survive these conditions remains unknown. Here we show that the integrated stress response (ISR) is uniquely active in HSCs and facilitates their persistence. Activating transcription factor 4 (ATF4) mediates the ISR and is highly expressed in HSCs due to scarcity of the eIF2 translation initiation complex. Amino acid deprivation results in eIF2α phosphorylation-dependent upregulation of ATF4, promoting HSC survival. Primitive acute myeloid leukemia (AML) cells also display eIF2 scarcity and ISR activity marks leukemia stem cells (LSCs) in primary AML samples. These findings identify a link between the ISR and stem cell survival in the normal and leukemic contexts.
Collapse
Affiliation(s)
- Peter van Galen
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nathan Mbong
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Antonia Kreso
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Erwin M Schoof
- The Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre (BRIC), Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Elvin Wagenblast
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Stanley W K Ng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5G 1A1, Canada
| | - Gabriela Krivdova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Liqing Jin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
13
|
Ye F, Huang J, Wang H, Luo C, Zhao K. Targeting epigenetic machinery: Emerging novel allosteric inhibitors. Pharmacol Ther 2019; 204:107406. [PMID: 31521697 DOI: 10.1016/j.pharmthera.2019.107406] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2019] [Indexed: 12/13/2022]
Abstract
Epigenetics has emerged as an extremely exciting fast-growing area of biomedical research in post genome era. Epigenetic dysfunction is tightly related with various diseases such as cancer and aging related degeneration, potentiating epigenetics modulators as important therapeutics targets. Indeed, inhibitors of histone deacetylase and DNA methyltransferase have been approved for treating blood tumor malignancies, whereas inhibitors of histone methyltransferase and histone acetyl-lysine recognizer bromodomain are in clinical stage. However, it remains a great challenge to discover potent and selective inhibitors by targeting catalytic site, as the same subfamily of epigenetic enzymes often share high sequence identity and very conserved catalytic core pocket. It is well known that epigenetic modifications are usually carried out by multi-protein complexes, and activation of catalytic subunit is often tightly regulated by other interactive protein component, especially in disease conditions. Therefore, it is not unusual that epigenetic complex machinery may exhibit allosteric regulation site induced by protein-protein interactions. Targeting allosteric site emerges as a compelling alternative strategy to develop epigenetic drugs with enhanced druggability and pharmacological profiles. In this review, we highlight recent progress in the development of allosteric inhibitors for epigenetic complexes through targeting protein-protein interactions. We also summarized the status of clinical applications of those inhibitors. Finally, we provide perspectives of future novel allosteric epigenetic machinery modulators emerging from otherwise undruggable single protein target.
Collapse
Affiliation(s)
- Fei Ye
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, China; College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jing Huang
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China; Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, China; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Cheng Luo
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China; Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, South Dong Qing Road, Guizhou 550025, China.
| | - Kehao Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, China; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| |
Collapse
|
14
|
Mutant H3 histones drive human pre-leukemic hematopoietic stem cell expansion and promote leukemic aggressiveness. Nat Commun 2019; 10:2891. [PMID: 31253791 PMCID: PMC6599207 DOI: 10.1038/s41467-019-10705-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 05/23/2019] [Indexed: 01/01/2023] Open
Abstract
Our ability to manage acute myeloid leukemia (AML) is limited by our incomplete understanding of the epigenetic disruption central to leukemogenesis, including improper histone methylation. Here we examine 16 histone H3 genes in 434 primary AML samples and identify Q69H, A26P, R2Q, R8H and K27M/I mutations (1.6%), with higher incidence in secondary AML (9%). These mutations occur in pre-leukemic hematopoietic stem cells (HSCs) and exist in the major leukemic clones in patients. They increase the frequency of functional HSCs, alter differentiation, and amplify leukemic aggressiveness. These effects are dependent on the specific mutation. H3K27 mutation increases the expression of genes involved in erythrocyte and myeloid differentiation with altered H3K27 tri-methylation and K27 acetylation. The functional impact of histone mutations is independent of RUNX1 mutation, although they at times co-occur. This study establishes that H3 mutations are drivers of human pre-cancerous stem cell expansion and important early events in leukemogenesis.
Collapse
|
15
|
Aqaqe N, Yassin M, Yassin AA, Ershaid N, Katz-Even C, Zipin-Roitman A, Kugler E, Lechman ER, Gan OI, Mitchell A, Dick JE, Izraeli S, Milyavsky M. An ERG Enhancer-Based Reporter Identifies Leukemia Cells with Elevated Leukemogenic Potential Driven by ERG-USP9X Feed-Forward Regulation. Cancer Res 2019; 79:3862-3876. [PMID: 31175119 DOI: 10.1158/0008-5472.can-18-3215] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/21/2019] [Accepted: 06/04/2019] [Indexed: 11/16/2022]
Abstract
Acute leukemia is a rapidly progressing blood cancer with low survival rates. Unfavorable prognosis is attributed to insufficiently characterized subpopulations of leukemia stem cells (LSC) that drive chemoresistance and leukemia relapse. Here we utilized a genetic reporter that assesses stemness to enrich and functionally characterize LSCs. We observed heterogeneous activity of the ERG+85 enhancer-based fluorescent reporter in human leukemias. Cells with high reporter activity (tagBFPHigh) exhibited elevated expression of stemness and chemoresistance genes and demonstrated increased clonogenicity and resistance to chemo- and radiotherapy as compared with their tagBFPNeg counterparts. The tagBFPHigh fraction was capable of regenerating the original cellular heterogeneity and demonstrated increased invasive ability. Moreover, the tagBFPHigh fraction was enriched for leukemia-initiating cells in a xenograft assay. We identified the ubiquitin hydrolase USP9X as a novel ERG transcriptional target that sustains ERG+85-positive cells by controlling ERG ubiquitination. Therapeutic targeting of USP9X led to preferential inhibition of the ERG-dependent leukemias. Collectively, these results characterize human leukemia cell functional heterogeneity and suggest that targeting ERG via USP9X inhibition may be a potential treatment strategy in patients with leukemia. SIGNIFICANCE: This study couples a novel experimental tool with state-of-the-art approaches to delineate molecular mechanisms underlying stem cell-related characteristics in leukemia cells.
Collapse
Affiliation(s)
- Nasma Aqaqe
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Muhammad Yassin
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abed Alkader Yassin
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nour Ershaid
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chen Katz-Even
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Zipin-Roitman
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eitan Kugler
- Department of Pediatric Hemato-Oncology, Schneider Children Medical Center Petah-Tikva, Israel.,The Gene Development and Environment Pediatric Research Institute, Pediatric Hemato-Oncology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Olga I Gan
- Princess Margaret Cancer Centre, University Health Network and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Amanda Mitchell
- Princess Margaret Cancer Centre, University Health Network and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Shai Izraeli
- Department of Pediatric Hemato-Oncology, Schneider Children Medical Center Petah-Tikva, Israel.,The Gene Development and Environment Pediatric Research Institute, Pediatric Hemato-Oncology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Milyavsky
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
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, Andreeff M. Mitochondrial ClpP-Mediated Proteolysis Induces Selective Cancer Cell Lethality. Cancer Cell 2019; 35:721-737.e9. [PMID: 31056398 PMCID: PMC6620028 DOI: 10.1016/j.ccell.2019.03.014] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/13/2018] [Accepted: 03/29/2019] [Indexed: 12/20/2022]
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.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Survival/drug effects
- Crystallography, X-Ray
- Drug Screening Assays, Antitumor
- Endopeptidase Clp/chemistry
- Endopeptidase Clp/genetics
- Endopeptidase Clp/metabolism
- Female
- HCT116 Cells
- HEK293 Cells
- Heterocyclic Compounds, 4 or More Rings/administration & dosage
- Heterocyclic Compounds, 4 or More Rings/chemistry
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Humans
- Imidazoles
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Mitochondria/metabolism
- Models, Molecular
- Point Mutation
- Protein Conformation/drug effects
- Proteolysis
- Pyridines
- Pyrimidines
- Tumor Suppressor Protein p53/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jo Ishizawa
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA
| | - Sarah F Zarabi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - R Eric Davis
- The University of Texas MD Anderson Cancer Center; Department of Lymphoma and Myeloma, Houston, TX 77030, USA
| | - Ondrej Halgas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Takenobu Nii
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA
| | - Yulia Jitkova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ran Zhao
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA
| | - Jonathan St-Germain
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Lauren E Heese
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA
| | - Grace Egan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Vivian R Ruvolo
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA
| | - Samir H Barghout
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Yuki Nishida
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Wencai Ma
- The University of Texas MD Anderson Cancer Center, Bioinformatics and Comp Biology, Houston, TX 77030, USA
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Todd Link
- The University of Texas MD Anderson Cancer Center, Genomic Medicine, Houston, TX 77030, USA
| | - Keith Wong
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mark Mabanglo
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kensuke Kojima
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA; Saga University, Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga 849-8501, Japan
| | - Gautam Borthakur
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA
| | - Neil MacLean
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Man Chun John Ma
- The University of Texas MD Anderson Cancer Center; Department of Lymphoma and Myeloma, Houston, TX 77030, USA
| | - Andrew B Leber
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Walid Houry
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Hagop Kantarjian
- The University of Texas MD Anderson Cancer Center; Department of Leukemia, Houston, TX 77030, USA
| | | | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Emil F Pai
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L7, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L7, Canada.
| | - Michael Andreeff
- The University of Texas MD Anderson Cancer Center, Molecular Hematology and Therapy, Department of Leukemia, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center; Department of Leukemia, Houston, TX 77030, USA.
| |
Collapse
|
17
|
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, Schimmer AD. The Mitochondrial Transacylase, Tafazzin, Regulates for AML Stemness by Modulating Intracellular Levels of Phospholipids. Cell Stem Cell 2019; 24:621-636.e16. [PMID: 30930145 DOI: 10.1016/j.stem.2019.02.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/19/2018] [Accepted: 02/27/2019] [Indexed: 12/17/2022]
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.
Collapse
Affiliation(s)
- Ayesh K Seneviratne
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mingjing Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Juan J Aristizabal Henao
- Laboratory of Nutritional Lipidomics, Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada
| | - Val A Fajardo
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Zhenyue Hao
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Veronique Voisin
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - G Wei Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - S Kim
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Neil MacLean
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xiaoming Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Danny Jeyaraju
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yulia Jitkova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Troy Ketela
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - David Sharon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Geethu Thomas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - James R Hawley
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Caitlin Schafer
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Helen Loo Yau
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Zaza Khuchua
- Department of Biochemistry, Sechenov Medical University, Moscow, Russian Federation; Institute of Medical Research Ilia State University, Tbilisi, Georgia
| | - Ahmed Aman
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, ON, Canada
| | - Rima Al-Awar
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, ON, Canada
| | - Atan Gross
- Department of Biological Regulation, Weizmann Institute, Rehovot, Israel
| | - Steven M Claypool
- Department of Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Steven Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ken D Stark
- Laboratory of Nutritional Lipidomics, Department of Kinesiology, University of Waterloo, Waterloo, ON, Canada
| | - Paul LeBlanc
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
18
|
Preclinical evaluation of the selective small-molecule UBA1 inhibitor, TAK-243, in acute myeloid leukemia. Leukemia 2018; 33:37-51. [DOI: 10.1038/s41375-018-0167-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/06/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023]
|
19
|
Roma A, Rota SG, Spagnuolo PA. Diosmetin Induces Apoptosis of Acute Myeloid Leukemia Cells. Mol Pharm 2018; 15:1353-1360. [DOI: 10.1021/acs.molpharmaceut.7b01151] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alessia Roma
- Department of Food Science, University of Guelph, 50 Stone Road E, Guelph, Ontario, Canada, N1G 2W1
| | - Sarah G. Rota
- Department of Food Science, University of Guelph, 50 Stone Road E, Guelph, Ontario, Canada, N1G 2W1
| | - Paul A. Spagnuolo
- Department of Food Science, University of Guelph, 50 Stone Road E, Guelph, Ontario, Canada, N1G 2W1
- School of Pharmacy, University of Waterloo, 10A Victoria Street S, Kitchener, Ontario, Canada, N2G 1C5
| |
Collapse
|
20
|
Staquicini DI, D'Angelo S, Ferrara F, Karjalainen K, Sharma G, Smith TL, Tarleton CA, Jaalouk DE, Kuniyasu A, Baze WB, Chaffee BK, Hanley PW, Barnhart KF, Koivunen E, Marchiò S, Sidman RL, Cortes JE, Kantarjian HM, Arap W, Pasqualini R. Therapeutic targeting of membrane-associated GRP78 in leukemia and lymphoma: preclinical efficacy in vitro and formal toxicity study of BMTP-78 in rodents and primates. THE PHARMACOGENOMICS JOURNAL 2017; 18:436-443. [PMID: 29205207 DOI: 10.1038/tpj.2017.46] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/19/2017] [Accepted: 06/30/2017] [Indexed: 01/11/2023]
Abstract
Translation of drug candidates into clinical settings requires demonstration of preclinical efficacy and formal toxicology analysis for filling an Investigational New Drug (IND) application with the US Food and Drug Administration (FDA). Here, we investigate the membrane-associated glucose response protein 78 (GRP78) as a therapeutic target in leukemia and lymphoma. We evaluated the efficacy of the GRP78-targeted proapoptotic drug bone metastasis targeting peptidomimetic 78 (BMTP-78), a member of the D(KLAKLAK)2-containing class of agents. BMTP-78 was validated in cells from patients with acute myeloid leukemia and in a panel of human leukemia and lymphoma cell lines, where it induced dose-dependent cytotoxicity in all samples tested. Based on the in vitro efficacy of BMTP-78, we performed formal good laboratory practice toxicology studies in both rodents (mice and rats) and nonhuman primates (cynomolgus and rhesus monkeys). These analyses represent required steps towards an IND application of BMTP-78 for theranostic first-in-human clinical trials.
Collapse
Affiliation(s)
- D I Staquicini
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - S D'Angelo
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - F Ferrara
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - K Karjalainen
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - G Sharma
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - T L Smith
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - C A Tarleton
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - D E Jaalouk
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - A Kuniyasu
- Department of Molecular Cell Pharmacology, Sojo University, Kumamoto, Japan
| | - W B Baze
- Department of Veterinary Science and Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - B K Chaffee
- Department of Veterinary Science and Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - P W Hanley
- Department of Veterinary Science and Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - K F Barnhart
- Department of Veterinary Science and Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA.,David H Koch Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - E Koivunen
- Department of Biological and Environmental Science, The University of Helsinki, Helsinki, Finland.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - S Marchiò
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA.,Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Center-FPO, IRCCS, Candiolo, Italy
| | - R L Sidman
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - J E Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - H M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W Arap
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico School of Medicine, Albuquerque, NM
| | - R Pasqualini
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.,Department of Internal Medicine, Division of Molecular Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
21
|
De Grandis M, Bardin F, Fauriat C, Zemmour C, El-Kaoutari A, Sergé A, Granjeaud S, Pouyet L, Montersino C, Chretien AS, Mozziconacci MJ, Castellano R, Bidaut G, Boher JM, Collette Y, Mancini SJC, Vey N, Aurrand-Lions M. JAM-C Identifies Src Family Kinase-Activated Leukemia-Initiating Cells and Predicts Poor Prognosis in Acute Myeloid Leukemia. Cancer Res 2017; 77:6627-6640. [PMID: 28972073 DOI: 10.1158/0008-5472.can-17-1223] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/24/2017] [Accepted: 09/25/2017] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) originates from hematopoietic stem and progenitor cells that acquire somatic mutations, leading to disease and clonogenic evolution. AML is characterized by accumulation of immature myeloid cells in the bone marrow and phenotypic cellular heterogeneity reflective of normal hematopoietic differentiation. Here, we show that JAM-C expression defines a subset of leukemic cells endowed with leukemia-initiating cell activity (LIC). Stratification of de novo AML patients at diagnosis based on JAM-C-expressing cells frequencies in the blood served as an independent prognostic marker for disease outcome. Using publicly available leukemic stem cell (LSC) gene expression profiles and gene expression data generated from JAM-C-expressing leukemic cells, we defined a single cell core gene expression signature correlated to JAM-C expression that reveals LSC heterogeneity. Finally, we demonstrated that JAM-C controls Src family kinase (SFK) activation in LSC and that LIC with exacerbated SFK activation was uniquely found within the JAM-C-expressing LSC compartment. Cancer Res; 77(23); 6627-40. ©2017 AACR.
Collapse
Affiliation(s)
- Maria De Grandis
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Florence Bardin
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Cyril Fauriat
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Christophe Zemmour
- Unité de Biostatistique et de Méthodologie, Département de la Recherche Clinique et de l'Innovation, Institut Paoli-Calmettes, Marseille, France
| | | | - Arnauld Sergé
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Samuel Granjeaud
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Laurent Pouyet
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Camille Montersino
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Anne-Sophie Chretien
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Marie-Joelle Mozziconacci
- Département de Biopathologie, Cytogénétique et Biologie Moléculaire, Institut Paoli-Calmettes, Marseille, France
| | - Remy Castellano
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Ghislain Bidaut
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Jean-Marie Boher
- Unité de Biostatistique et de Méthodologie, Département de la Recherche Clinique et de l'Innovation, Institut Paoli-Calmettes, Marseille, France
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Marseille, France
| | - Yves Collette
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Stéphane J C Mancini
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Norbert Vey
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
- Département d'Hématologie, Institut Paoli-Calmettes, Marseille, France
| | - Michel Aurrand-Lions
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.
| |
Collapse
|
22
|
Zipin-Roitman A, Aqaqe N, Yassin M, Biechonski S, Amar M, van Delft MF, Gan OI, McDermott SP, Buzina A, Ketela T, Shlush L, Xie S, Voisin V, Moffat J, Minden MD, Dick JE, Milyavsky M. SMYD2 lysine methyltransferase regulates leukemia cell growth and regeneration after genotoxic stress. Oncotarget 2017; 8:16712-16727. [PMID: 28187429 PMCID: PMC5369996 DOI: 10.18632/oncotarget.15147] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/24/2017] [Indexed: 12/12/2022] Open
Abstract
The molecular determinants governing escape of Acute Myeloid Leukemia (AML) cells from DNA damaging therapy remain poorly defined and account for therapy failures. To isolate genes responsible for leukemia cells regeneration following multiple challenges with irradiation we performed a genome-wide shRNA screen. Some of the isolated hits are known players in the DNA damage response (e.g. p53, CHK2), whereas other, e.g. SMYD2 lysine methyltransferase (KMT), remains uncharacterized in the AML context. Here we report that SMYD2 knockdown confers relative resistance to human AML cells against multiple classes of DNA damaging agents. Induction of the transient quiescence state upon SMYD2 downregulation correlated with the resistance. We revealed that diminished SMYD2 expression resulted in the upregulation of the related methyltransferase SET7/9, suggesting compensatory relationships. Indeed, pharmacological targeting of SET7/9 with (R)-PFI2 inhibitor preferentially inhibited the growth of cells expressing low levels of SMYD2. Finally, decreased expression of SMYD2 in AML patients correlated with the reduced sensitivity to therapy and lower probability to achieve complete remission. We propose that the interplay between SMYD2 and SET7/9 levels shifts leukemia cells from growth to quiescence state that is associated with the higher resistance to DNA damaging agents and rationalize SET7/9 pharmacological targeting in AML.
Collapse
Affiliation(s)
- Adi Zipin-Roitman
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nasma Aqaqe
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Muhammad Yassin
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shahar Biechonski
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mariam Amar
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mark F van Delft
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada.,Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Olga I Gan
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Sean P McDermott
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada.,Leidos Biomedical Research, Washington D.C., USA
| | - Alla Buzina
- Donnelly Centre for Cellular and Biomedical Research, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Troy Ketela
- Donnelly Centre for Cellular and Biomedical Research, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Liran Shlush
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Stephanie Xie
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Veronique Voisin
- Donnelly Centre for Cellular and Biomedical Research, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Jason Moffat
- Donnelly Centre for Cellular and Biomedical Research, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Michael Milyavsky
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
23
|
Rota SG, Roma A, Dude I, Ma C, Stevens R, MacEachern J, Graczyk J, Espiritu SMG, Rao PN, Minden MD, Kreinin E, Hess DA, Doxey AC, Spagnuolo PA. Estrogen Receptor β Is a Novel Target in Acute Myeloid Leukemia. Mol Cancer Ther 2017; 16:2618-2626. [DOI: 10.1158/1535-7163.mct-17-0292] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/11/2017] [Accepted: 08/16/2017] [Indexed: 11/16/2022]
|
24
|
Leveraging increased cytoplasmic nucleoside kinase activity to target mtDNA and oxidative phosphorylation in AML. Blood 2017; 129:2657-2666. [PMID: 28283480 DOI: 10.1182/blood-2016-10-741207] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/06/2017] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial DNA (mtDNA) biosynthesis requires replication factors and adequate nucleotide pools from the mitochondria and cytoplasm. We performed gene expression profiling analysis of 542 human acute myeloid leukemia (AML) samples and identified 55% with upregulated mtDNA biosynthesis pathway expression compared with normal hematopoietic cells. Genes that support mitochondrial nucleotide pools, including mitochondrial nucleotide transporters and a subset of cytoplasmic nucleoside kinases, were also increased in AML compared with normal hematopoietic samples. Knockdown of cytoplasmic nucleoside kinases reduced mtDNA levels in AML cells, demonstrating their contribution in maintaining mtDNA. To assess cytoplasmic nucleoside kinase pathway activity, we used a nucleoside analog 2'3'-dideoxycytidine (ddC), which is phosphorylated to the activated antimetabolite, 2'3'-dideoxycytidine triphosphate by cytoplasmic nucleoside kinases. ddC is a selective inhibitor of the mitochondrial DNA polymerase γ. ddC was preferentially activated in AML cells compared with normal hematopoietic progenitor cells. ddC treatment inhibited mtDNA replication, oxidative phosphorylation, and induced cytotoxicity in a panel of AML cell lines. Furthermore, ddC preferentially inhibited mtDNA replication in a subset of primary human leukemia cells and selectively targeted leukemia cells while sparing normal progenitor cells. In animal models of human AML, treatment with ddC decreased mtDNA, electron transport chain proteins, and induced tumor regression without toxicity. ddC also targeted leukemic stem cells in secondary AML xenotransplantation assays. Thus, AML cells have increased cytidine nucleoside kinase activity that regulates mtDNA biogenesis and can be leveraged to selectively target oxidative phosphorylation in AML.
Collapse
|
25
|
Rotin LE, Gronda M, MacLean N, Hurren R, Wang X, Lin FH, Wrana J, Datti A, Barber DL, Minden MD, Slassi M, Schimmer AD. Ibrutinib synergizes with poly(ADP-ribose) glycohydrolase inhibitors to induce cell death in AML cells via a BTK-independent mechanism. Oncotarget 2016; 7:2765-79. [PMID: 26624983 PMCID: PMC4823070 DOI: 10.18632/oncotarget.6409] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/16/2015] [Indexed: 12/15/2022] Open
Abstract
Targeting Bruton's tyrosine kinase (BTK) with the small molecule BTK inhibitor ibrutinib has significantly improved patient outcomes in several B-cell malignancies, with minimal toxicity. Given the reported expression and constitutive activation of BTK in acute myeloid leukemia (AML) cells, there has been recent interest in investigating the anti-AML activity of ibrutinib. We noted that ibrutinib had limited single-agent toxicity in a panel of AML cell lines and primary AML samples, and therefore sought to identify ibrutinib-sensitizing drugs. Using a high-throughput combination chemical screen, we identified that the poly(ADP-ribose) glycohydrolase (PARG) inhibitor ethacridine lactate synergized with ibrutinib in TEX and OCI-AML2 leukemia cell lines. The combination of ibrutinib and ethacridine induced a synergistic increase in reactive oxygen species that was functionally important to explain the observed cell death. Interestingly, synergistic cytotoxicity of ibrutinib and ethacridine was independent of the inhibitory effect of ibrutinib against BTK, as knockdown of BTK did not sensitize TEX and OCI-AML2 cells to ethacridine treatment. Thus, our findings indicate that ibrutinib may have a BTK-independent role in AML and that PARG inhibitors may have utility as part of a combination therapy for this disease.
Collapse
Affiliation(s)
- Lianne E Rotin
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Neil MacLean
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - XiaoMing Wang
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Feng-Hsu Lin
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Jeff Wrana
- Samuel Lunenfeld Research Institute, Toronto, Ontario, Canada
| | - Alessandro Datti
- Samuel Lunenfeld Research Institute, Toronto, Ontario, Canada.,Department of Agricultural, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Dwayne L Barber
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Tcheng M, Samudio I, Lee EA, Minden MD, Spagnuolo PA. The mitochondria target drug avocatin B synergizes with induction chemotherapeutics to induce leukemia cell death. Leuk Lymphoma 2016; 58:986-988. [DOI: 10.1080/10428194.2016.1218005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Bernard D, Gebbia M, Prabha S, Gronda M, MacLean N, Wang X, Hurren R, Sukhai MA, Cho EE, Manolson MF, Datti A, Wrana J, Minden MD, Al-Awar R, Aman A, Nislow C, Giaever G, Schimmer AD. Select microtubule inhibitors increase lysosome acidity and promote lysosomal disruption in acute myeloid leukemia (AML) cells. Apoptosis 2016; 20:948-59. [PMID: 25832785 DOI: 10.1007/s10495-015-1123-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
To identify new biological vulnerabilities in acute myeloid leukemia, we screened a library of natural products for compounds cytotoxic to TEX leukemia cells. This screen identified the novel small molecule Deoxysappanone B 7,4' dimethyl ether (Deox B 7,4), which possessed nanomolar anti-leukemic activity. To determine the anti-leukemic mechanism of action of Deox B 7,4, we conducted a genome-wide screen in Saccharomyces cerevisiae and identified enrichment of genes related to mitotic cell cycle as well as vacuolar acidification, therefore pointing to microtubules and vacuolar (V)-ATPase as potential drug targets. Further investigations into the mechanisms of action of Deox B 7,4 and a related analogue revealed that these compounds were reversible microtubule inhibitors that bound near the colchicine site. In addition, Deox B 7,4 and its analogue increased lysosomal V-ATPase activity and lysosome acidity. The effects on microtubules and lysosomes were functionally important for the anti-leukemic effects of these drugs. The lysosomal effects were characteristic of select microtubule inhibitors as only the Deox compounds and nocodazole, but not colchicine, vinca alkaloids or paclitaxel, altered lysosome acidity and induced lysosomal disruption. Thus, our data highlight a new mechanism of action of select microtubule inhibitors on lysosomal function.
Collapse
Affiliation(s)
- Dannie Bernard
- Princess Margaret Cancer Centre, University Health Network, Rm 9-516, 610 University Ave, Toronto, ON, M5G 2M9, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Nin DS, Li F, Visvanathan S, Khan M. Misfolded N-CoR is Linked to the Ectopic Reactivation of CD34/Flt3-Based Stem-Cell Phenotype in Promyelocytic and Monocytic Acute Myeloid Leukemia. Front Oncol 2015; 5:210. [PMID: 26500885 PMCID: PMC4595783 DOI: 10.3389/fonc.2015.00210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 09/14/2015] [Indexed: 12/15/2022] Open
Abstract
Nuclear receptor co-repressor (N-CoR) is the key component of generic co-repressor complex essential for the transcriptional control of genes involved in cellular hemostasis. We have recently reported that N-CoR actively represses Flt3, a key factor of hematopoietic stem cells (HSC) self-renewal and growth, and that de-repression of Flt3 by the misfolded N-CoR plays an important role in the pathogenesis of promyelocytic and monocytic acute myeloid leukemia (AML). The leukemic cells derived from the promyelocytic and monocytic AML are distinctly characterized by the ectopic reactivation of stem cell phenotypes in relatively committed myeloid compartment. However, the molecular mechanism underlying this phenomenon is not known. Here, we report that N-CoR function is essential for the commitment of primitive hematopoietic cells to the cells of myeloid lineage and that loss of N-CoR function due to misfolding is linked to the ectopic reactivation of generic stem cell phenotypes in promyelocytic and monocytic AML. Analysis of N-CoR and Flt3 transcripts in mouse hematopoietic cells revealed a positive correlation between N-CoR level and the commitment of myeloid cells and an inverse correlation between N-CoR and Flt3 levels in primitive as well as committed myeloid cells. Enforced N-CoR expression in mouse HSCs inhibited their growth and self-renewal potentials and promoted maturation toward cells of myeloid lineage, suggesting a role of N-CoR in the commitment of cells of myeloid lineage. In contrast to AML cells with natively folded N-CoR, primary and secondary promyelocytic and monocytic AML cells harboring the misfolded N-CoR were highly positive for Flt3 and myeloid antigen-based HSC marker CD34. Genetic and therapeutic restoration of N-CoR conformation significantly down-regulated the CD34 levels in monocytic AML cells, suggesting an important role of N-CoR in the suppression of CD34-based HSC phenotypes. These findings collectively suggest that N-CoR is crucial for the commitment of primitive hematopoietic cells to cells of myeloid lineage and that misfolded N-CoR may contribute to transformation of committed myeloid cells through the ectopic reactivation of Flt3/CD34-based stem cell phenotypes in promyelocytic and monocytic AML. Moreover, these findings provide novel mechanistic insights into the formation of leukemic stem cells in subsets of AML and identify the misfolded N-CoR as a subtype-specific biomarker of AML.
Collapse
Affiliation(s)
- Dawn Sijin Nin
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Feng Li
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore , Singapore
| | - Sridevi Visvanathan
- Department of Biochemistry, School of Medicine, AIMST University , Semeling , Malaysia
| | - Matiullah Khan
- Department of Pathology, School of Medicine, AIMST University , Semeling , Malaysia
| |
Collapse
|
29
|
Lee EA, Angka L, Rota SG, Hanlon T, Mitchell A, Hurren R, Wang XM, Gronda M, Boyaci E, Bojko B, Minden M, Sriskanthadevan S, Datti A, Wrana JL, Edginton A, Pawliszyn J, Joseph JW, Quadrilatero J, Schimmer AD, Spagnuolo PA. Targeting Mitochondria with Avocatin B Induces Selective Leukemia Cell Death. Cancer Res 2015; 75:2478-88. [PMID: 26077472 DOI: 10.1158/0008-5472.can-14-2676] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Treatment regimens for acute myeloid leukemia (AML) continue to offer weak clinical outcomes. Through a high-throughput cell-based screen, we identified avocatin B, a lipid derived from avocado fruit, as a novel compound with cytotoxic activity in AML. Avocatin B reduced human primary AML cell viability without effect on normal peripheral blood stem cells. Functional stem cell assays demonstrated selectivity toward AML progenitor and stem cells without effects on normal hematopoietic stem cells. Mechanistic investigations indicated that cytotoxicity relied on mitochondrial localization, as cells lacking functional mitochondria or CPT1, the enzyme that facilitates mitochondria lipid transport, were insensitive to avocatin B. Furthermore, avocatin B inhibited fatty acid oxidation and decreased NADPH levels, resulting in ROS-dependent leukemia cell death characterized by the release of mitochondrial proteins, apoptosis-inducing factor, and cytochrome c. This study reveals a novel strategy for selective leukemia cell eradication based on a specific difference in mitochondrial function.
Collapse
Affiliation(s)
- Eric A Lee
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Leonard Angka
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Sarah-Grace Rota
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Thomas Hanlon
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Andrew Mitchell
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - Rose Hurren
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Xiao Ming Wang
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Ezel Boyaci
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Barbara Bojko
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Mark Minden
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | | | - Alessandro Datti
- SMART Laboratory for High-Throughput Screening Programs, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada. Department of Agricultural, Food and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Jeffery L Wrana
- SMART Laboratory for High-Throughput Screening Programs, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Andrea Edginton
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Jamie W Joseph
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Center, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Paul A Spagnuolo
- School of Pharmacy, University of Waterloo, Kitchener, Ontario, Canada.
| |
Collapse
|
30
|
Kempema AM, Widen JC, Hexum JK, Andrews TE, Wang D, Rathe SK, Meece FA, Noble KE, Sachs Z, Largaespada DA, Harki DA. Synthesis and antileukemic activities of C1-C10-modified parthenolide analogues. Bioorg Med Chem 2015; 23:4737-4745. [PMID: 26088334 DOI: 10.1016/j.bmc.2015.05.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 05/24/2015] [Indexed: 01/08/2023]
Abstract
Parthenolide (PTL) is a sesquiterpene lactone natural product with anti-proliferative activity to cancer cells. Selective eradication of leukemic stem cells (LSCs) over healthy hematopoietic stem cells (HSCs) by PTL has been demonstrated in previous studies, which suggests PTL and related molecules may be useful for targeting LSCs. Eradication of LSCs is required for curative therapy. Chemical optimizations of PTL to improve potency and pharmacokinetic parameters have focused largely on the α-methylene-γ-butyrolactone, which is essential for activity. Conversely, we evaluated modifications to the C1-C10 olefin and benchmarked new inhibitors to PTL with respect to inhibitory potency across a panel of cancer cell lines, ability to target drug-resistant acute myeloid leukemia (AML) cells, efficacy for inhibiting clonal growth of AML cells, toxicity to healthy bone marrow cells, and efficiency for promoting intracellular reactive oxygen species (ROS) levels. Cyclopropane 4 was found to possess less toxicity to healthy bone marrow cells, enhanced potency for the induction of cellular ROS, and similar broad-spectrum anti-proliferative activity to cancer cells in comparison to PTL.
Collapse
Affiliation(s)
- Aaron M Kempema
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - John C Widen
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph K Hexum
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Timothy E Andrews
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dan Wang
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Susan K Rathe
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Frederick A Meece
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Klara E Noble
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zohar Sachs
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - David A Largaespada
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
31
|
Xenograft models for normal and malignant stem cells. Blood 2015; 125:2630-40. [DOI: 10.1182/blood-2014-11-570218] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/04/2015] [Indexed: 12/18/2022] Open
Abstract
Abstract
The model systems available for studying human hematopoiesis, malignant hematopoiesis, and hematopoietic stem cell (HSC) function in vivo have improved dramatically over the last decade, primarily due to improvements in xenograft mouse strains. Several recent reviews have focused on the historic development of immunodeficient mice over the last 2 decades, as well as their use in understanding human HSC and leukemia stem cell (LSC) biology and function in the context of a humanized mouse. However, in the intervening time since these reviews, a number of new mouse models, technical approaches, and scientific advances have been made. In this review, we update the reader on the newest and best models and approaches available for studying human malignant and normal HSCs in immunodeficient mice, including newly developed mice for use in chemotherapy testing and improved techniques for humanizing mice without laborious purification of HSC. We also review some relevant scientific findings from xenograft studies and highlight the continued limitations that confront researchers working with human HSC and LSC in vivo.
Collapse
|
32
|
ELMO1 is upregulated in AML CD34+ stem/progenitor cells, mediates chemotaxis and predicts poor prognosis in normal karyotype AML. PLoS One 2014; 9:e111568. [PMID: 25360637 PMCID: PMC4216115 DOI: 10.1371/journal.pone.0111568] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/03/2014] [Indexed: 12/27/2022] Open
Abstract
Both normal as well leukemic hematopoietic stem cells critically depend on their microenvironment in the bone marrow for processes such as self-renewal, survival and differentiation, although the exact pathways that are involved remain poorly understood. We performed transcriptome analysis on primitive CD34+ acute myeloid leukemia (AML) cells (n = 46), their more differentiated CD34- leukemic progeny, and normal CD34+ bone marrow cells (n = 31) and focused on differentially expressed genes involved in adhesion and migration. Thus, Engulfment and Motility protein 1 (ELMO1) was identified amongst the top 50 most differentially expressed genes. ELMO1 is a crucial link in the signaling cascade that leads to activation of RAC GTPases and cytoskeleton rearrangements. We confirmed increased ELMO1 expression at the mRNA and protein level in a panel of AML samples and showed that high ELMO1 expression is an independent negative prognostic factor in normal karyotype (NK) AML in three large independent patient cohorts. Downmodulation of ELMO1 in human CB CD34+ cells did not significantly alter expansion, progenitor frequency or differentiation in stromal co-cultures, but did result in a decreased frequency of stem cells in LTC-IC assays. In BCR-ABL-transduced human CB CD34+ cells depletion of ELMO1 resulted in a mild decrease in proliferation, but replating capacity of progenitors was severely impaired. Downregulation of ELMO1 in a panel of primary CD34+ AML cells also resulted in reduced long-term growth in stromal co-cultures in two out of three cases. Pharmacological inhibition of the ELMO1 downstream target RAC resulted in a severely impaired proliferation and survival of leukemic cells. Finally, ELMO1 depletion caused a marked decrease in SDF1-induced chemotaxis of leukemic cells. Taken together, these data show that inhibiting the ELMO1-RAC axis might be an alternative way to target leukemic cells.
Collapse
|
33
|
Tursky ML, Beck D, Thoms JAI, Huang Y, Kumari A, Unnikrishnan A, Knezevic K, Evans K, Richards LA, Lee E, Morris J, Goldberg L, Izraeli S, Wong JWH, Olivier J, Lock RB, MacKenzie KL, Pimanda JE. Overexpression of ERG in cord blood progenitors promotes expansion and recapitulates molecular signatures of high ERG leukemias. Leukemia 2014; 29:819-27. [PMID: 25306899 DOI: 10.1038/leu.2014.299] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 09/04/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023]
Abstract
High expression of the ETS family transcription factor ERG is associated with poor clinical outcome in acute myeloid leukemia (AML) and acute T-cell lymphoblastic leukemia (T-ALL). In murine models, high ERG expression induces both T-ALL and AML. However, no study to date has defined the effect of high ERG expression on primary human hematopoietic cells. In the present study, human CD34+ cells were transduced with retroviral vectors to elevate ERG gene expression to levels detected in high ERG AML. RNA sequencing was performed on purified populations of transduced cells to define the effects of high ERG on gene expression in human CD34+ cells. Integration of the genome-wide expression data with other data sets revealed that high ERG drives an expression signature that shares features of normal hematopoietic stem cells, high ERG AMLs, early T-cell precursor-ALLs and leukemic stem cell signatures associated with poor clinical outcome. Functional assays linked this gene expression profile to enhanced progenitor cell expansion. These results support a model whereby a stem cell gene expression network driven by high ERG in human cells enhances the expansion of the progenitor pool, providing opportunity for the acquisition and propagation of mutations and the development of leukemia.
Collapse
Affiliation(s)
- M L Tursky
- 1] Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW, Sydney, Australia [2] Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - D Beck
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - J A I Thoms
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - Y Huang
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - A Kumari
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - A Unnikrishnan
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - K Knezevic
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - K Evans
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - L A Richards
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - E Lee
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - J Morris
- Department of Obstetrics and Gynaecology, Royal North Shore Hospital, University of Sydney, Sydney, Australia
| | - L Goldberg
- 1] Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel [2] Department of Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, Israel
| | - S Izraeli
- 1] Cancer Research Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel [2] Department of Human Molecular Genetics and Biochemistry, Tel Aviv University, Tel Aviv, Israel
| | - J W H Wong
- Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - J Olivier
- School of Mathematics and Statistics, UNSW, Sydney, Australia
| | - R B Lock
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - K L MacKenzie
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - J E Pimanda
- 1] Adult Cancer Program, Prince of Wales Clinical School, Lowy Cancer Research Centre, UNSW, Sydney, Australia [2] Department of Haematology, Prince of Wales Hospital, Sydney, Australia
| |
Collapse
|
34
|
Gedye CA, Hussain A, Paterson J, Smrke A, Saini H, Sirskyj D, Pereira K, Lobo N, Stewart J, Go C, Ho J, Medrano M, Hyatt E, Yuan J, Lauriault S, Meyer M, Kondratyev M, van den Beucken T, Jewett M, Dirks P, Guidos CJ, Danska J, Wang J, Wouters B, Neel B, Rottapel R, Ailles LE. Cell surface profiling using high-throughput flow cytometry: a platform for biomarker discovery and analysis of cellular heterogeneity. PLoS One 2014; 9:e105602. [PMID: 25170899 PMCID: PMC4149490 DOI: 10.1371/journal.pone.0105602] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/22/2014] [Indexed: 11/18/2022] Open
Abstract
Cell surface proteins have a wide range of biological functions, and are often used as lineage-specific markers. Antibodies that recognize cell surface antigens are widely used as research tools, diagnostic markers, and even therapeutic agents. The ability to obtain broad cell surface protein profiles would thus be of great value in a wide range of fields. There are however currently few available methods for high-throughput analysis of large numbers of cell surface proteins. We describe here a high-throughput flow cytometry (HT-FC) platform for rapid analysis of 363 cell surface antigens. Here we demonstrate that HT-FC provides reproducible results, and use the platform to identify cell surface antigens that are influenced by common cell preparation methods. We show that multiple populations within complex samples such as primary tumors can be simultaneously analyzed by co-staining of cells with lineage-specific antibodies, allowing unprecedented depth of analysis of heterogeneous cell populations. Furthermore, standard informatics methods can be used to visualize, cluster and downsample HT-FC data to reveal novel signatures and biomarkers. We show that the cell surface profile provides sufficient molecular information to classify samples from different cancers and tissue types into biologically relevant clusters using unsupervised hierarchical clustering. Finally, we describe the identification of a candidate lineage marker and its subsequent validation. In summary, HT-FC combines the advantages of a high-throughput screen with a detection method that is sensitive, quantitative, highly reproducible, and allows in-depth analysis of heterogeneous samples. The use of commercially available antibodies means that high quality reagents are immediately available for follow-up studies. HT-FC has a wide range of applications, including biomarker discovery, molecular classification of cancers, or identification of novel lineage specific or stem cell markers.
Collapse
Affiliation(s)
- Craig A Gedye
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ali Hussain
- Dept. of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Joshua Paterson
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Alannah Smrke
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Harleen Saini
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Danylo Sirskyj
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Keira Pereira
- Dept. of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Nazleen Lobo
- Dept. of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jocelyn Stewart
- Dept. of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Christopher Go
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jenny Ho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mauricio Medrano
- Dept. of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Elzbieta Hyatt
- Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Julie Yuan
- Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Stevan Lauriault
- Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | | | - Maria Kondratyev
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Michael Jewett
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Peter Dirks
- Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Cynthia J Guidos
- Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Jayne Danska
- Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Jean Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Bradly Wouters
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Dept. of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Benjamin Neel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Dept. of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Robert Rottapel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Laurie E Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Dept. of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
35
|
van Galen P, Kreso A, Wienholds E, Laurenti E, Eppert K, Lechman ER, Mbong N, Hermans K, Dobson S, April C, Fan JB, Dick JE. Reduced lymphoid lineage priming promotes human hematopoietic stem cell expansion. Cell Stem Cell 2014; 14:94-106. [PMID: 24388174 DOI: 10.1016/j.stem.2013.11.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 10/07/2013] [Accepted: 11/25/2013] [Indexed: 01/10/2023]
Abstract
The hematopoietic system sustains regeneration throughout life by balancing self-renewal and differentiation. To stay poised for mature blood production, hematopoietic stem cells (HSCs) maintain low-level expression of lineage-associated genes, a process termed lineage priming. Here, we modulated expression levels of Inhibitor of DNA binding (ID) proteins to ask whether lineage priming affects self-renewal of human HSCs. We found that lentiviral overexpression of ID proteins in cord blood HSCs biases myeloerythroid commitment at the expense of lymphoid differentiation. Conversely, reducing ID2 expression levels increases lymphoid potential. Mechanistically, ID2 inhibits the transcription factor E47 to attenuate B-lymphoid priming in HSCs and progenitors. Strikingly, ID2 overexpression also results in a 10-fold expansion of HSCs in serial limiting dilution assays, indicating that early lymphoid transcription factors antagonize human HSC self-renewal. The relationship between lineage priming and self-renewal can be exploited to increase expansion of transplantable human HSCs and points to broader implications for other stem cell populations.
Collapse
Affiliation(s)
- Peter van Galen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Antonija Kreso
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Erno Wienholds
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Elisa Laurenti
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kolja Eppert
- Department of Pediatrics, McGill University and the Research Institute of the McGill University Health Centre, Westmount, QC H3Z 2Z3, Canada
| | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Nathan Mbong
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Karin Hermans
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Stephanie Dobson
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | | | | | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
36
|
Abstract
Serial sampling of the cellular composition of bone marrow (BM) is a routine procedure critical to clinical hematology. This protocol describes a detailed step-by-step technical procedure for an analogous procedure in live mice which allows for serial characterization of cells present in the BM. This procedure facilitates studies aimed to detect the presence of exogenously administered cells within the BM of mice as would be done in xenograft studies for instance. Moreover, this procedure allows for the retrieval and characterization of cells enriched in the BM such as hematopoietic stem and progenitor cells (HSPCs) without sacrifice of mice. Given that the cellular composition of peripheral blood is not necessarily reflective of proportions and types of stem and progenitor cells present in the marrow, procedures which provide access to this compartment without requiring termination of the mice are very helpful. The use of femoral bone marrow aspiration is illustrated here for cytological analysis of marrow cells, flow cytometric characterization of the hematopoietic stem/progenitor compartment, and culture of sorted HSPCs obtained by femoral BM aspiration compared with conventional marrow harvest.
Collapse
Affiliation(s)
- Young Rock Chung
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center
| | - Eunhee Kim
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center;
| |
Collapse
|
37
|
Jitkova Y, Gronda M, Hurren R, Wang X, Goard CA, Jhas B, Schimmer AD. A novel formulation of tigecycline has enhanced stability and sustained antibacterial and antileukemic activity. PLoS One 2014; 9:e95281. [PMID: 24871339 PMCID: PMC4037176 DOI: 10.1371/journal.pone.0095281] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 03/25/2014] [Indexed: 01/23/2023] Open
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.
Collapse
Affiliation(s)
- Yulia Jitkova
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Xiaoming Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Carolyn A. Goard
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Bozhena Jhas
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Aaron D. Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- * E-mail:
| |
Collapse
|
38
|
van Galen P, Kreso A, Mbong N, Kent DG, Fitzmaurice T, Chambers JE, Xie S, Laurenti E, Hermans K, Eppert K, Marciniak SJ, Goodall JC, Green AR, Wouters BG, Wienholds E, Dick JE. The unfolded protein response governs integrity of the haematopoietic stem-cell pool during stress. Nature 2014; 510:268-72. [DOI: 10.1038/nature13228] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 03/05/2014] [Indexed: 01/08/2023]
|
39
|
Peltekova VD, Lemire M, Qazi AM, Zaidi SHE, Trinh QM, Bielecki R, Rogers M, Hodgson L, Wang M, D'Souza DJA, Zandi S, Chong T, Kwan JYY, Kozak K, De Borja R, Timms L, Rangrej J, Volar M, Chan-Seng-Yue M, Beck T, Ash C, Lee S, Wang J, Boutros PC, Stein LD, Dick JE, Gryfe R, McPherson JD, Zanke BW, Pollett A, Gallinger S, Hudson TJ. Identification of genes expressed by immune cells of the colon that are regulated by colorectal cancer-associated variants. Int J Cancer 2013; 134:2330-41. [PMID: 24154973 PMCID: PMC3949167 DOI: 10.1002/ijc.28557] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 09/27/2013] [Indexed: 12/19/2022]
Abstract
A locus on human chromosome 11q23 tagged by marker rs3802842 was associated with colorectal cancer (CRC) in a genome-wide association study; this finding has been replicated in case–control studies worldwide. In order to identify biologic factors at this locus that are related to the etiopathology of CRC, we used microarray-based target selection methods, coupled to next-generation sequencing, to study 103 kb at the 11q23 locus. We genotyped 369 putative variants from 1,030 patients with CRC (cases) and 1,061 individuals without CRC (controls) from the Ontario Familial Colorectal Cancer Registry. Two previously uncharacterized genes, COLCA1 and COLCA2, were found to be co-regulated genes that are transcribed from opposite strands. Expression levels of COLCA1 and COLCA2 transcripts correlate with rs3802842 genotypes. In colon tissues, COLCA1 co-localizes with crystalloid granules of eosinophils and granular organelles of mast cells, neutrophils, macrophages, dendritic cells and differentiated myeloid-derived cell lines. COLCA2 is present in the cytoplasm of normal epithelial, immune and other cell lineages, as well as tumor cells. Tissue microarray analysis demonstrates the association of rs3802842 with lymphocyte density in the lamina propria (p = 0.014) and levels of COLCA1 in the lamina propria (p = 0.00016) and COLCA2 (tumor cells, p = 0.0041 and lamina propria, p = 6 × 10–5). In conclusion, genetic, expression and immunohistochemical data implicate COLCA1 and COLCA2 in the pathogenesis of colon cancer. Histologic analyses indicate the involvement of immune pathways.
Collapse
|
40
|
Yu W, Chory EJ, Wernimont AK, Tempel W, Scopton A, Federation A, Marineau JJ, Qi J, Barsyte-Lovejoy D, Yi J, Marcellus R, Iacob RE, Engen JR, Griffin C, Aman A, Wienholds E, Li F, Pineda J, Estiu G, Shatseva T, Hajian T, Al-Awar R, Dick JE, Vedadi M, Brown PJ, Arrowsmith CH, Bradner JE, Schapira M. Catalytic site remodelling of the DOT1L methyltransferase by selective inhibitors. Nat Commun 2013; 3:1288. [PMID: 23250418 DOI: 10.1038/ncomms2304] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 11/15/2012] [Indexed: 01/04/2023] Open
Abstract
Selective inhibition of protein methyltransferases is a promising new approach to drug discovery. An attractive strategy towards this goal is the development of compounds that selectively inhibit binding of the cofactor, S-adenosylmethionine, within specific protein methyltransferases. Here we report the three-dimensional structure of the protein methyltransferase DOT1L bound to EPZ004777, the first S-adenosylmethionine-competitive inhibitor of a protein methyltransferase with in vivo efficacy. This structure and those of four new analogues reveal remodelling of the catalytic site. EPZ004777 and a brominated analogue, SGC0946, inhibit DOT1L in vitro and selectively kill mixed lineage leukaemia cells, in which DOT1L is aberrantly localized via interaction with an oncogenic MLL fusion protein. These data provide important new insight into mechanisms of cell-active S-adenosylmethionine-competitive protein methyltransferase inhibitors, and establish a foundation for the further development of drug-like inhibitors of DOT1L for cancer therapy.
Collapse
Affiliation(s)
- Wenyu Yu
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Jhas B, Sriskanthadevan S, Skrtic M, Sukhai MA, Voisin V, Jitkova Y, Gronda M, Hurren R, Laister RC, Bader GD, Minden MD, Schimmer AD. Metabolic adaptation to chronic inhibition of mitochondrial protein synthesis in acute myeloid leukemia cells. PLoS One 2013; 8:e58367. [PMID: 23520503 PMCID: PMC3592803 DOI: 10.1371/journal.pone.0058367] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 02/04/2013] [Indexed: 12/19/2022] Open
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.
Collapse
MESH Headings
- Anti-Bacterial Agents/pharmacology
- Cell Line, Tumor
- Drug Resistance, Neoplasm
- Electron Transport Complex IV/biosynthesis
- 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/metabolism
- Leukemia, Myeloid, Acute/pathology
- Minocycline/analogs & derivatives
- Minocycline/pharmacology
- Mitochondrial Proteins/biosynthesis
- Mitochondrial Proteins/genetics
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Oxygen Consumption/drug effects
- Oxygen Consumption/genetics
- Protein Biosynthesis
- Tigecycline
Collapse
Affiliation(s)
- Bozhena Jhas
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Shrivani Sriskanthadevan
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Marko Skrtic
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Mahadeo A. Sukhai
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | | | - Yulia Jitkova
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Marcela Gronda
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Rose Hurren
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Rob C. Laister
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Gary D. Bader
- The Donnelly Centre, University of Toronto, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Mark D. Minden
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Aaron D. Schimmer
- The Princess Margaret Hospital and The Ontario Cancer Institute, University Health Network, Toronto, Canada
- * E-mail:
| |
Collapse
|
42
|
Sukhai MA, Prabha S, Hurren R, Rutledge AC, Lee AY, Sriskanthadevan S, Sun H, Wang X, Skrtic M, Seneviratne A, Cusimano M, Jhas B, Gronda M, MacLean N, Cho EE, Spagnuolo PA, Sharmeen S, Gebbia M, Urbanus M, Eppert K, Dissanayake D, Jonet A, Dassonville-Klimpt A, Li X, Datti A, Ohashi PS, Wrana J, Rogers I, Sonnet P, Ellis WY, Corey SJ, Eaves C, Minden MD, Wang JC, Dick JE, Nislow C, Giaever G, Schimmer AD. Lysosomal disruption preferentially targets acute myeloid leukemia cells and progenitors. J Clin Invest 2013; 123:315-28. [PMID: 23202731 PMCID: PMC3533286 DOI: 10.1172/jci64180] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 10/04/2012] [Indexed: 01/15/2023] Open
Abstract
Despite efforts to understand and treat acute myeloid leukemia (AML), there remains a need for more comprehensive therapies to prevent AML-associated relapses. To identify new therapeutic strategies for AML, we screened a library of on- and off-patent drugs and identified the antimalarial agent mefloquine as a compound that selectively kills AML cells and AML stem cells in a panel of leukemia cell lines and in mice. Using a yeast genome-wide functional screen for mefloquine sensitizers, we identified genes associated with the yeast vacuole, the homolog of the mammalian lysosome. Consistent with this, we determined that mefloquine disrupts lysosomes, directly permeabilizes the lysosome membrane, and releases cathepsins into the cytosol. Knockdown of the lysosomal membrane proteins LAMP1 and LAMP2 resulted in decreased cell viability, as did treatment of AML cells with known lysosome disrupters. Highlighting a potential therapeutic rationale for this strategy, leukemic cells had significantly larger lysosomes compared with normal cells, and leukemia-initiating cells overexpressed lysosomal biogenesis genes. These results demonstrate that lysosomal disruption preferentially targets AML cells and AML progenitor cells, providing a rationale for testing lysosomal disruption as a novel therapeutic strategy for AML.
Collapse
Affiliation(s)
- Mahadeo A. Sukhai
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Swayam Prabha
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rose Hurren
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Angela C. Rutledge
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anna Y. Lee
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shrivani Sriskanthadevan
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hong Sun
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xiaoming Wang
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marko Skrtic
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ayesh Seneviratne
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Maria Cusimano
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Bozhena Jhas
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marcela Gronda
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Neil MacLean
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Eunice E. Cho
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Paul A. Spagnuolo
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sumaiya Sharmeen
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marinella Gebbia
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Malene Urbanus
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kolja Eppert
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dilan Dissanayake
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alexia Jonet
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alexandra Dassonville-Klimpt
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xiaoming Li
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alessandro Datti
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Pamela S. Ohashi
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jeff Wrana
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ian Rogers
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Pascal Sonnet
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - William Y. Ellis
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Seth J. Corey
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Connie Eaves
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mark D. Minden
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean C.Y. Wang
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - John E. Dick
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Corey Nislow
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Guri Giaever
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Aaron D. Schimmer
- Princess Margaret Hospital/the Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada.
Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada.
Laboratoire des Glucides, CNRS FRE 3517, UFR de Pharmacie, Université de Picardie Jules Verne, 1, Amiens, France.
Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.
Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy.
Department of Chemical Informatics, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.
Departments of Pediatrics and Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Shahab S, Shamsi TS, Ahmed N. Molecular Involvement and Prognostic Importance of Fms-like Tyrosine Kinase 3 in Acute Myeloid Leukemia. Asian Pac J Cancer Prev 2012; 13:4215-20. [DOI: 10.7314/apjcp.2012.13.8.4215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
44
|
Schimmer AD, Skrtić M. Therapeutic potential of mitochondrial translation inhibition for treatment of acute myeloid leukemia. Expert Rev Hematol 2012; 5:117-9. [PMID: 22475277 DOI: 10.1586/ehm.12.8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Leukemia-associated antigens and their relevance to the immunotherapy of acute myeloid leukemia. Leukemia 2012; 26:2186-96. [PMID: 22652755 DOI: 10.1038/leu.2012.145] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The graft-versus-leukemia effect of allogeneic hematopoietic stem cell transplantation (HSCT) has shown that the immune system is capable of eradicating acute myeloid leukemia (AML). This knowledge, along with the identification of the target antigens against which antileukemia immune responses are directed, has provided a strong impetus for the development of antigen-targeted immunotherapy of AML. The success of any antigen-specific immunotherapeutic strategy depends critically on the choice of target antigen. Ideal molecules for immune targeting in AML are those that are: (1) leukemia-specific; (2) expressed in most leukemic blasts including leukemic stem cells; (3) important for the leukemic phenotype; (4) immunogenic; and (5) clinically effective. In this review, we provide a comprehensive overview on AML-related tumor antigens and assess their applicability for immunotherapy against the five criteria outlined above. In this way, we aim to facilitate the selection of appropriate target antigens, a task that has become increasingly challenging given the large number of antigens identified and the rapid pace at which new targets are being discovered. The information provided in this review is intended to guide the rational design of future antigen-specific immunotherapy trials, which will hopefully lead to new antileukemia therapies with more selectivity and higher efficacy.
Collapse
|
46
|
Bae J, Song W, Smith R, Daley J, Tai YT, Anderson KC, Munshi NC. A novel immunogenic CS1-specific peptide inducing antigen-specific cytotoxic T lymphocytes targeting multiple myeloma. Br J Haematol 2012; 157:687-701. [PMID: 22533610 DOI: 10.1111/j.1365-2141.2012.09111.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/15/2012] [Indexed: 01/19/2023]
Abstract
The CS1 antigen provides a unique target for the development of an immunotherapeutic strategy to treat patients with multiple myeloma (MM). This study aimed to identify HLA-A2(+) immunogenic peptides from the CS1 antigen, which induce peptide-specific cytotoxic T lymphocytes (CTL) against HLA-A2(+) MM cells. We identified a novel immunogenic HLA-A2-specific CS1(239-247) (SLFVLGLFL) peptide, which induced CS1-specific CTL (CS1-CTL) to MM cells. The CS1-CTL showed a distinct phenotype, with an increased percentage of effector memory and activated CTL and a decreased percentage of naïve CTL. CS1(239-247) peptide-specific CD8(+) T cells were detected by DimerX analyses and demonstrated functional activities specific to the peptide. The CTL displayed HLA-A2-restricted and antigen-specific cytotoxicity, proliferation, degranulation and γ-interferon (IFN-γ) production against both primary MM cells and MM cell lines. In addition, the effector memory cells subset (CD45RO(+) CCR7(-) /CD3(+) CD8(+) ) within CS1-CTL showed a higher level of CD107a degranulation and IFN-γ production as compared to effector cells (CD45RO(-) CCR7(-) /CD3(+) CD8(+) ) against HLA-A2(+) primary MM cells or MM cell lines. In conclusion, this study introduced a novel immunogenic HLA-A2-specific CS1(239-247) peptide capable of inducing antigen-specific CTL against MM cells that will provide a framework for its application as a novel MM immunotherapy.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Schenk T, Chen WC, Göllner S, Howell L, Jin L, Hebestreit K, Klein HU, Popescu AC, Burnett A, Mills K, Casero RA, Marton L, Woster P, Minden MD, Dugas M, Wang JCY, Dick JE, Müller-Tidow C, Petrie K, Zelent A. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med 2012; 18:605-11. [PMID: 22406747 DOI: 10.1038/nm.2661] [Citation(s) in RCA: 518] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 01/03/2012] [Indexed: 12/14/2022]
Abstract
Acute promyelocytic leukemia (APL), a cytogenetically distinct subtype of acute myeloid leukemia (AML), characterized by the t(15;17)-associated PML-RARA fusion, has been successfully treated with therapy utilizing all-trans-retinoic acid (ATRA) to differentiate leukemic blasts. However, among patients with non-APL AML, ATRA-based treatment has not been effective. Here we show that, through epigenetic reprogramming, inhibitors of lysine-specific demethylase 1 (LSD1, also called KDM1A), including tranylcypromine (TCP), unlocked the ATRA-driven therapeutic response in non-APL AML. LSD1 inhibition did not lead to a large-scale increase in histone 3 Lys4 dimethylation (H3K4(me2)) across the genome, but it did increase H3K4(me2) and expression of myeloid-differentiation-associated genes. Notably, treatment with ATRA plus TCP markedly diminished the engraftment of primary human AML cells in vivo in nonobese diabetic (NOD)-severe combined immunodeficient (SCID) mice, suggesting that ATRA in combination with TCP may target leukemia-initiating cells. Furthermore, initiation of ATRA plus TCP treatment 15 d after engraftment of human AML cells in NOD-SCID γ (with interleukin-2 (IL-2) receptor γ chain deficiency) mice also revealed the ATRA plus TCP drug combination to have a potent anti-leukemic effect that was superior to treatment with either drug alone. These data identify LSD1 as a therapeutic target and strongly suggest that it may contribute to AML pathogenesis by inhibiting the normal pro-differentiative function of ATRA, paving the way for new combinatorial therapies for AML.
Collapse
Affiliation(s)
- Tino Schenk
- Division of Molecular Pathology, Institute of Cancer Research, Sutton, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
A small molecule screening strategy with validation on human leukemia stem cells uncovers the therapeutic efficacy of kinetin riboside. Blood 2012; 119:1200-7. [DOI: 10.1182/blood-2011-01-330019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Abstract
Gene regulatory networks that govern hematopoietic stem cells (HSCs) and leukemia-initiating cells (L-ICs) are deeply entangled. Thus, the discovery of compounds that target L-ICs while sparing HSC is an attractive but difficult endeavor. Presently, most screening approaches fail to counter-screen compounds against normal hematopoietic stem/progenitor cells (HSPCs). Here, we present a multistep in vitro and in vivo approach to identify compounds that can target L-ICs in acute myeloid leukemia (AML). A high-throughput screen of 4000 compounds on novel leukemia cell lines derived from human experimental leukemogenesis models yielded 80 hits, of which 10 were less toxic to HSPC. We characterized a single compound, kinetin riboside (KR), on AML L-ICs and HSPCs. KR demonstrated comparable efficacy to standard therapies against blast cells in 63 primary leukemias. In vitro, KR targeted the L-IC–enriched CD34+CD38− AML fraction, while sparing HSPC-enriched fractions, although these effects were mitigated on HSC assayed in vivo. KR eliminated L-ICs in 2 of 4 primary AML samples when assayed in vivo and highlights the importance of in vivo L-IC and HSC assays to measure function. Overall, we provide a novel approach to screen large drug libraries for the discovery of anti–L-IC compounds for human leukemias.
Collapse
|
49
|
Škrtić 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, Schimmer AD. Inhibition of mitochondrial translation as a therapeutic strategy for human acute myeloid leukemia. Cancer Cell 2011; 20:674-88. [PMID: 22094260 PMCID: PMC3221282 DOI: 10.1016/j.ccr.2011.10.015] [Citation(s) in RCA: 523] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 09/05/2011] [Accepted: 10/14/2011] [Indexed: 12/17/2022]
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.
Collapse
Affiliation(s)
- Marko Škrtić
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Shrivani Sriskanthadevan
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Bozhena Jhas
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Marinella Gebbia
- Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S 3E1 Canada
| | - Xiaoming Wang
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Zezhou Wang
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Rose Hurren
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Yulia Jitkova
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Marcela Gronda
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Neil Maclean
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Courteney K. Lai
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, V5Z 1L3 Canada
| | - Yanina Eberhard
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Justyna Bartoszko
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Paul Spagnuolo
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Angela C. Rutledge
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Alessandro Datti
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5 Canada
| | - Troy Ketela
- Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S 3E1 Canada
| | - Jason Moffat
- Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S 3E1 Canada
| | - Brian H. Robinson
- Genetics and Genome Biology, The Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 1X8 Canada
| | - Jessie H. Cameron
- Genetics and Genome Biology, The Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 1X8 Canada
| | - Jeffery Wrana
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5 Canada
| | - Connie J. Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, V5Z 1L3 Canada
| | - Mark D. Minden
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
| | - Jean C.Y. Wang
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
- Division of Stem Cell and Developmental Biology, Campbell Family Institute for Cancer Research/Ontario Cancer Institute, Toronto, Ontario M5G 1L7, Canada
| | - John E. Dick
- Division of Stem Cell and Developmental Biology, Campbell Family Institute for Cancer Research/Ontario Cancer Institute, Toronto, Ontario M5G 1L7, Canada
| | - Keith Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, V5Z 1L3 Canada
| | - Corey Nislow
- Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S 3E1 Canada
| | - Guri Giaever
- Department of Molecular Genetics, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S 3E1 Canada
| | - Aaron D. Schimmer
- The Campbell Family Cancer Research Institute, The Princess Margaret Hospital, The Ontario Cancer Institute, Toronto, ON, M5G 2M9 Canada
- To whom correspondence should be addressed: Aaron D. Schimmer, Princess Margaret Hospital, Rm 9-516, 610 University Ave, Toronto, ON, Canada M5G 2M9, Tel: 416-946-2838, Fax: 416-946-6546,
| |
Collapse
|
50
|
Waldron T, De Dominici M, Soliera AR, Audia A, Iacobucci I, Lonetti A, Martinelli G, Zhang Y, Martinez R, Hyslop T, Bender TP, Calabretta B. c-Myb and its target Bmi1 are required for p190BCR/ABL leukemogenesis in mouse and human cells. Leukemia 2011; 26:644-53. [PMID: 21960247 PMCID: PMC3252490 DOI: 10.1038/leu.2011.264] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Expression of c-Myb is required for normal hematopoiesis and for proliferation of myeloid leukemia blasts and a subset of T cell leukemia but its role in B-cell leukemogenesis is unknown. We tested the role of c-Myb in p190BCR/ABL-dependent B-cell leukemia in mice transplanted with p190BCR/ABL-transduced marrow cells with a c-Myb allele (Mybf/d) and in double transgenic p190BCR/ABL/Mybw/d mice. In both models, loss of a c-Myb allele caused a less aggressive B-cell leukemia. In p190BCR/ABL expressing human B-cell leukemia lines, knockdown of c-Myb expression suppressed proliferation and colony formation. Compared to c-Mybw/f cells, expression of Bmi1, a regulator of stem cell proliferation and maintenance, was decreased in pre-B cells from Mybw/d p190BCR/ABL transgenic mice. Ectopic expression of a mutant c-Myb or Bmi1 enhanced the proliferation and colony formation of Mybw/d p190BCR/ABL B-cells; by contrast, Bmi1 downregulation inhibited colony formation of p190BCR/ABL-expressing murine B cells and human B-cell leukemia lines. Moreover, c-Myb interacted with a segment of the human Bmi1 promoter and enhanced its activity. In blasts from nineteen Ph1 adult ALL patients, levels of c-Myb and Bmi1 showed a positive correlation. Together, these findings support the existence of a c-Myb-Bmi1 transcription regulatory pathway required for p190BCR/ABL leukemogenesis.
Collapse
Affiliation(s)
- T Waldron
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|