251
|
Aldehyde biphenyl chalcones induce immunogenic apoptotic-like cell death and are promising new safe compounds against a wide range of hematologic cancers. Future Med Chem 2020; 12:673-688. [PMID: 32191531 DOI: 10.4155/fmc-2019-0228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Investigate the apoptotic mechanisms of two new aldehyde biphenyl chalcones on leukemia cells. Materials & methods: From a series of 71 new chalcones, we selected the two most cytotoxic. Results: JA3 and JA7 were cytotoxic not only against hematological malignancies but also against solid tumor and cancer stem cells, yet with no toxicity to normal cells. Moreover, they induced immunogenic apoptotic-like cell death independently of promyelocytic leukemia protein, with extensive mitochondrial damages downstream of endoplasmic reticulum stress. Preventing endoplasmic reticulum stress and the upregulation of proapoptotic machinery inhibited JA3- and JA7-induced cell death. Likewise, blocking receptor Fas protected cells from killing. They increased the antileukemic effect of cytarabine and vincristine and killed leukemic cells collected from patients with different acute leukemia subtypes. Conclusion: JA3 and JA7 represent new promising prototypes for the development of new chemotherapeutics.
Collapse
|
252
|
Huang H, Weng H, Deng X, Chen J. RNA Modifications in Cancer: Functions, Mechanisms, and Therapeutic Implications. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2020. [DOI: 10.1146/annurev-cancerbio-030419-033357] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Over 170 chemical modifications have been identified in protein-coding and noncoding RNAs and shown to exhibit broad impacts on gene expression. Dysregulation of RNA modifications caused by aberrant expression of or mutations in RNA modifiers aberrantly reprograms the epitranscriptome and skews global gene expression, which in turn leads to tumorigenesis and drug resistance. Here we review current knowledge of the functions and underlying mechanisms of aberrant RNA modifications in human cancers, particularly several common RNA modifications, including N6-methyladenosine (m6A), A-to-I editing, pseudouridine (ψ), 5-methylcytosine (m5C), 5-hydroxymethylcytosine (hm5C), N1-methyladenosine (m1A), and N4-acetylcytidine (ac4C), providing insights into therapeutic implications of targeting RNA modifications and the associated machineries for cancer therapy.
Collapse
Affiliation(s)
- Huilin Huang
- Department of Systems Biology and The Gehr Family Center for Leukemia Research, The Beckman Research Institute of City of Hope, Monrovia, California 91010, USA
| | - Hengyou Weng
- Department of Systems Biology and The Gehr Family Center for Leukemia Research, The Beckman Research Institute of City of Hope, Monrovia, California 91010, USA
| | - Xiaolan Deng
- Department of Systems Biology and The Gehr Family Center for Leukemia Research, The Beckman Research Institute of City of Hope, Monrovia, California 91010, USA
| | - Jianjun Chen
- Department of Systems Biology and The Gehr Family Center for Leukemia Research, The Beckman Research Institute of City of Hope, Monrovia, California 91010, USA
| |
Collapse
|
253
|
Knorr KLB, Goldberg AD. Leukemia stem cell gene expression signatures contribute to acute myeloid leukemia risk stratification. Haematologica 2020; 105:533-536. [PMID: 32115413 DOI: 10.3324/haematol.2019.241117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Katherine L B Knorr
- Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aaron D Goldberg
- Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
254
|
Brown MA, Edwards MA, Alshiraihi I, Geng H, Dekker JD, Tucker HO. The lysine methyltransferase SMYD2 is required for normal lymphocyte development and survival of hematopoietic leukemias. Genes Immun 2020; 21:119-130. [PMID: 32115575 PMCID: PMC7183909 DOI: 10.1038/s41435-020-0094-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022]
Abstract
The 5 membered SET and MYND Domain-containing lysine methyltransferase (SMYD) family plays pivotal roles in development and proliferation. Initially characterized within the cardiovascular system, one such member, SMYD2, has been implicated as an oncogene in leukemias deriving from flawed hematopoietic stem cell (HSC) differentiation. We show here that conditional SMYD2 loss disrupts hematopoiesis at and downstream of the HSC via both apoptotic loss and transcriptional deregulation of HSC proliferation and disruption of Wnt-β-Catenin signaling. Yet previously documented SMYD2 cell cycle targets were unscathed. Turning our analysis to human leukemias, we observed that SMYD2 is highly expressed in CML, MLLr-B-ALL, AML, T-ALL and B-ALL leukemias and its levels in B-ALL correlate with poor survival. SMYD2 knockdown results in apoptotic death and loss of anchorage-independent transformation of each of these hematopoietic leukemias. These data provide an underlying mechanism by which SMYD2 acts during normal hematopoiesis and as a proto-oncogene in leukemia.
Collapse
Affiliation(s)
- Mark A Brown
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.,Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, 80523, USA
| | - Melissa A Edwards
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, 80523, USA.,Department of Molecular Biosciences, The University of Texas at Austin, 1 University Station A5000, Austin, TX, 78712, USA
| | - Ilham Alshiraihi
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, 80523, USA
| | - Huimin Geng
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Joseph D Dekker
- Department of Molecular Biosciences, The University of Texas at Austin, 1 University Station A5000, Austin, TX, 78712, USA
| | - Haley O Tucker
- Department of Molecular Biosciences, The University of Texas at Austin, 1 University Station A5000, Austin, TX, 78712, USA.
| |
Collapse
|
255
|
Hatzl S, Perfler B, Wurm S, Uhl B, Quehenberger F, Ebner S, Troppmair J, Reinisch A, Wölfler A, Sill H, Zebisch A. Increased Expression of Micro-RNA-23a Mediates Chemoresistance to Cytarabine in Acute Myeloid Leukemia. Cancers (Basel) 2020; 12:496. [PMID: 32093419 PMCID: PMC7072365 DOI: 10.3390/cancers12020496] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022] Open
Abstract
Resistance to chemotherapy is one of the primary obstacles in acute myeloid leukemia (AML) therapy. Micro-RNA-23a (miR-23a) is frequently deregulated in AML and has been linked to chemoresistance in solid cancers. We, therefore, studied its role in chemoresistance to cytarabine (AraC), which forms the backbone of all cytostatic AML treatments. Initially, we assessed AraC sensitivity in three AML cell lines following miR-23a overexpression/knockdown using MTT-cell viability and soft-agar colony-formation assays. Overexpression of miR-23a decreased the sensitivity to AraC, whereas its knockdown had the opposite effect. Analysis of clinical data revealed that high miR-23a expression correlated with relapsed/refractory (R/R) AML disease stages, the leukemic stem cell compartment, as well as with inferior overall survival (OS) and event-free survival (EFS) in AraC-treated patients. Mechanistically, we demonstrate that miR-23a targets and downregulates topoisomerase-2-beta (TOP2B), and that TOP2B knockdown mediates AraC chemoresistance as well. Likewise, low TOP2B expression also correlated with R/R-AML disease stages and inferior EFS/OS. In conclusion, we show that increased expression of miR-23a mediates chemoresistance to AraC in AML and that it correlates with an inferior outcome in AraC-treated AML patients. We further demonstrate that miR-23a causes the downregulation of TOP2B, which is likely to mediate its effects on AraC sensitivity.
Collapse
Affiliation(s)
- Stefan Hatzl
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (S.H.); (B.P.); (S.W.); (B.U.); (A.R.); (A.W.); (H.S.)
| | - Bianca Perfler
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (S.H.); (B.P.); (S.W.); (B.U.); (A.R.); (A.W.); (H.S.)
| | - Sonja Wurm
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (S.H.); (B.P.); (S.W.); (B.U.); (A.R.); (A.W.); (H.S.)
| | - Barbara Uhl
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (S.H.); (B.P.); (S.W.); (B.U.); (A.R.); (A.W.); (H.S.)
| | - Franz Quehenberger
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, 8036 Graz, Austria;
| | - Susanne Ebner
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.E.); (J.T.)
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.E.); (J.T.)
| | - Andreas Reinisch
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (S.H.); (B.P.); (S.W.); (B.U.); (A.R.); (A.W.); (H.S.)
| | - Albert Wölfler
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (S.H.); (B.P.); (S.W.); (B.U.); (A.R.); (A.W.); (H.S.)
| | - Heinz Sill
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (S.H.); (B.P.); (S.W.); (B.U.); (A.R.); (A.W.); (H.S.)
| | - Armin Zebisch
- Division of Hematology, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria; (S.H.); (B.P.); (S.W.); (B.U.); (A.R.); (A.W.); (H.S.)
- Otto-Loewi-Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010 Graz, Austria
| |
Collapse
|
256
|
Sheng Y, Yu C, Liu Y, Hu C, Ma R, Lu X, Ji P, Chen J, Mizukawa B, Huang Y, Licht JD, Qian Z. FOXM1 regulates leukemia stem cell quiescence and survival in MLL-rearranged AML. Nat Commun 2020; 11:928. [PMID: 32066721 PMCID: PMC7026046 DOI: 10.1038/s41467-020-14590-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 12/16/2019] [Indexed: 12/16/2022] Open
Abstract
FOXM1, a known transcription factor, promotes cell proliferation in a variety of cancer cells. Here we show that Foxm1 is required for survival, quiescence and self-renewal of MLL-AF9 (MA9)-transformed leukemia stem cells (LSCs) in vivo. Mechanistically, Foxm1 upregulation activates the Wnt/β-catenin signaling pathways by directly binding to β-catenin and stabilizing β-catenin protein through inhibiting its degradation, thereby preserving LSC quiescence, and promoting LSC self-renewal in MLL-rearranged AML. More importantly, inhibition of FOXM1 markedly suppresses leukemogenic potential and induces apoptosis of primary LSCs from MLL-rearranged AML patients in vitro and in vivo in xenograft mice. Thus, our study shows a critical role and mechanisms of Foxm1 in MA9-LSCs, and indicates that FOXM1 is a potential therapeutic target for selectively eliminating LSCs in MLL-rearranged AML.
Collapse
Affiliation(s)
- Yue Sheng
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Chunjie Yu
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yin Liu
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Chao Hu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Rui Ma
- Institute for Tuberculosis Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Xinyan Lu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, IL, USA
| | - Peng Ji
- Department of Pathology, Feinberg School of Medicine, Northwestern University, IL, USA
| | - Jianjun Chen
- Department of System Biology, City of Hope, CA, USA
| | - Benjamin Mizukawa
- Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yong Huang
- Department of Medicine, University of Virginia, Charlottestville, VA, USA
| | - Jonathan D Licht
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Zhijian Qian
- Division of Hematology/Oncology, UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
257
|
Abstract
Acute megakaryoblastic leukemia (AMKL) is a rare malignancy affecting megakaryocytes, platelet-producing cells that reside in the bone marrow. Children with Down syndrome (DS) are particularly prone to developing the disease and have a different age of onset, distinct genetic mutations, and better prognosis as compared with individuals without DS who develop the disease. Here, we discuss the contributions of chromosome 21 genes and other genetic mutations to AMKL, the clinical features of the disease, and the differing features of DS- and non-DS-AMKL. Further studies elucidating the role of chromosome 21 genes in this disease may aid our understanding of how they function in other types of leukemia, in which they are frequently mutated or differentially expressed. Although researchers have made many insights into understanding AMKL, much more remains to be learned about its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Maureen McNulty
- Northwestern University, Division of Hematology/Oncology, Chicago, Illinois 60611, USA
| | - John D Crispino
- Northwestern University, Division of Hematology/Oncology, Chicago, Illinois 60611, USA
| |
Collapse
|
258
|
Chao MP, Takimoto CH, Feng DD, McKenna K, Gip P, Liu J, Volkmer JP, Weissman IL, Majeti R. Therapeutic Targeting of the Macrophage Immune Checkpoint CD47 in Myeloid Malignancies. Front Oncol 2020; 9:1380. [PMID: 32038992 PMCID: PMC6990910 DOI: 10.3389/fonc.2019.01380] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/22/2019] [Indexed: 02/04/2023] Open
Abstract
In recent years, immunotherapies have been clinically investigated in AML and other myeloid malignancies. While most of these are focused on stimulating the adaptive immune system (including T cell checkpoint inhibitors), several key approaches targeting the innate immune system have been identified. Macrophages are a key cell type in the innate immune response with CD47 being identified as a dominant macrophage checkpoint. CD47 is a "do not eat me" signal, overexpressed in myeloid malignancies that leads to tumor evasion of phagocytosis by macrophages. Blockade of CD47 leads to engulfment of leukemic cells and therapeutic elimination. Pre-clinical data has demonstrated robust anti-cancer activity in multiple hematologic malignancies including AML and myelodysplastic syndrome (MDS). In addition, clinical studies have been underway with CD47 targeting agents in both AML and MDS as monotherapy and in combination. This review will describe the role of CD47 in myeloid malignancies and pre-clinical data supporting CD47 targeting. In addition, initial clinical data of CD47 targeting in AML/MDS will be reviewed, and including the first-in-class anti-CD47 antibody magrolimab.
Collapse
Affiliation(s)
- Mark P Chao
- Forty Seven, Inc., Menlo Park, CA, United States
| | | | | | | | - Phung Gip
- Forty Seven, Inc., Menlo Park, CA, United States
| | - Jie Liu
- Forty Seven, Inc., Menlo Park, CA, United States
| | | | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, CA, United States
| | - Ravindra Majeti
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, CA, United States.,Division of Hematology, Department of Medicine, Stanford, CA, United States
| |
Collapse
|
259
|
Qasrawi A, Gomes V, Chacko CA, Mansour A, Kesler M, Arora R, Wei S, Ramlal R, Munker R. Acute undifferentiated leukemia: data on incidence and outcomes from a large population-based database. Leuk Res 2020; 89:106301. [PMID: 31982153 DOI: 10.1016/j.leukres.2020.106301] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 02/03/2023]
Abstract
Acute undifferentiated leukemia (AUL) is rare and defined by the absence of bona fide myeloid and lymphoid markers. Little is known about its incidence, survival and optimal management in the recent time period. Based on a case observed in our clinic, we queried the Surveillance, Epidemiology, and End Results database between 2000 and 2016. A total of 1,888 cases of AUL were diagnosed (1.34 per million person-years). The incidence of AUL has significantly decreased over time. Compared to other acute leukemias, patients with AUL have the highest median age (74 years); in contrast to acute myeloid leukemia (AML, 65) and acute lymphoblastic leukemia (ALL, 12). Excluding patients with preexisting malignancies, 1,444 patients with AUL were analyzed for survival. Only 35% of AUL patients had received chemotherapy. Comparatively, 94% of ALL and 71% of AML cases received chemotherapy. Among AUL patients who received chemotherapy, the median survival was 12 months as opposed to 1 month in the group who did not receive chemotherapy (or unknown status). Among adults, AUL patients had the worst prognosis, with a median overall survival (OS) of 9 months, compared to 27 months in ALL and 13 months in AML. Among children, the median OS was superior for all three groups of leukemias, the OS of AUL patients being better than in AML and very similar to ALL. On multivariate analysis, older age and time period were associated with worse outcome. We describe here the largest series of cases with AUL published to date.
Collapse
Affiliation(s)
- Ayman Qasrawi
- Department of Medicine, University of Kentucky, Lexington, KY, USA
| | | | | | - Akila Mansour
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Melissa Kesler
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Ranjana Arora
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Sainan Wei
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Reshma Ramlal
- Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Reinhold Munker
- Department of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
260
|
Ha YN, Song S, Orlikova-Boyer B, Cerella C, Christov C, Kijjoa A, Diederich M. Petromurin C Induces Protective Autophagy and Apoptosis in FLT3-ITD-Positive AML: Synergy with Gilteritinib. Mar Drugs 2020; 18:md18010057. [PMID: 31963113 PMCID: PMC7024157 DOI: 10.3390/md18010057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/25/2022] Open
Abstract
Treatment of acute myeloid leukemia (AML) remains inefficient due to drug resistance and relapse, particularly in patients with FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD). Marine-derived natural products have recently been used for drug development against AML. We show in this study that petromurin C, which was isolated from the culture extract of the marine-derived fungus Aspergillus candidus KUFA0062, isolated from the marine sponge Epipolasis sp., induces early autophagy followed by apoptotic cell death via activation of the intrinsic cell death pathway concomitant with mitochondrial stress and downregulation of Mcl-1 in FLT3-ITD mutated MV4-11 cells. Moreover, petromurin C synergized with the clinically-used FLT3 inhibitor gilteritinib at sub-toxic concentrations. Altogether, our results provide preliminary indications that petromurin C provides anti-leukemic effects alone or in combination with gilteritinib.
Collapse
MESH Headings
- Aniline Compounds/administration & dosage
- Aniline Compounds/pharmacology
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis/drug effects
- Aquatic Organisms/chemistry
- Autophagy/drug effects
- Biological Products/administration & dosage
- Biological Products/pharmacology
- Cell Line, Tumor
- Down-Regulation/drug effects
- Drug Resistance, Neoplasm
- Drug Synergism
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Pyrazines/administration & dosage
- Pyrazines/pharmacology
- Signal Transduction/drug effects
- U937 Cells
- Zebrafish
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- You Na Ha
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Korea; (Y.N.H.); (S.S.)
| | - Sungmi Song
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Korea; (Y.N.H.); (S.S.)
| | - Barbora Orlikova-Boyer
- Laboratoire de Biologie Moléculaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg; (B.O.-B.); (C.C.)
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg; (B.O.-B.); (C.C.)
| | - Christo Christov
- Service d’Histologie, Faculté de Médicine, Université de Lorraine, INSERM U1256 NGERE, 54000 Nancy, France;
| | - Anake Kijjoa
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto de Lexões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08626, Korea; (Y.N.H.); (S.S.)
- Correspondence: ; Tel.: +82-2-880-8919
| |
Collapse
|
261
|
Mitchell K, Steidl U. Targeting Immunophenotypic Markers on Leukemic Stem Cells: How Lessons from Current Approaches and Advances in the Leukemia Stem Cell (LSC) Model Can Inform Better Strategies for Treating Acute Myeloid Leukemia (AML). Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036251. [PMID: 31451539 DOI: 10.1101/cshperspect.a036251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Therapies targeting cell-surface antigens in acute myeloid leukemia (AML) have been tested over the past 20 years with limited improvement in overall survival. Recent advances in the understanding of AML pathogenesis support therapeutic targeting of leukemia stem cells as the most promising avenue toward a cure. In this review, we provide an overview of the evolving leukemia stem cell (LSC) model, including evidence of the cell of origin, cellular and molecular disease architecture, and source of relapse in AML. In addition, we explore limitations of current targeted strategies utilized in AML and describe the various immunophenotypic antigens that have been proposed as LSC-directed therapeutic targets. We draw lessons from current approaches as well as from the (pre)-LSC model to suggest criteria that immunophenotypic targets should meet for more specific and effective elimination of disease-initiating clones, highlighting in detail a few targets that we suggest fit these criteria most completely.
Collapse
Affiliation(s)
- Kelly Mitchell
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, New York 10461, USA.,Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
262
|
MacPherson L, Anokye J, Yeung MM, Lam EYN, Chan YC, Weng CF, Yeh P, Knezevic K, Butler MS, Hoegl A, Chan KL, Burr ML, Gearing LJ, Willson T, Liu J, Choi J, Yang Y, Bilardi RA, Falk H, Nguyen N, Stupple PA, Peat TS, Zhang M, de Silva M, Carrasco-Pozo C, Avery VM, Khoo PS, Dolezal O, Dennis ML, Nuttall S, Surjadi R, Newman J, Ren B, Leaver DJ, Sun Y, Baell JB, Dovey O, Vassiliou GS, Grebien F, Dawson SJ, Street IP, Monahan BJ, Burns CJ, Choudhary C, Blewitt ME, Voss AK, Thomas T, Dawson MA. HBO1 is required for the maintenance of leukaemia stem cells. Nature 2020; 577:266-270. [PMID: 31827282 DOI: 10.1038/s41586-019-1835-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/12/2019] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by transcriptional dysregulation that results in a block in differentiation and increased malignant self-renewal. Various epigenetic therapies aimed at reversing these hallmarks of AML have progressed into clinical trials, but most show only modest efficacy owing to an inability to effectively eradicate leukaemia stem cells (LSCs)1. Here, to specifically identify novel dependencies in LSCs, we screened a bespoke library of small hairpin RNAs that target chromatin regulators in a unique ex vivo mouse model of LSCs. We identify the MYST acetyltransferase HBO1 (also known as KAT7 or MYST2) and several known members of the HBO1 protein complex as critical regulators of LSC maintenance. Using CRISPR domain screening and quantitative mass spectrometry, we identified the histone acetyltransferase domain of HBO1 as being essential in the acetylation of histone H3 at K14. H3 acetylated at K14 (H3K14ac) facilitates the processivity of RNA polymerase II to maintain the high expression of key genes (including Hoxa9 and Hoxa10) that help to sustain the functional properties of LSCs. To leverage this dependency therapeutically, we developed a highly potent small-molecule inhibitor of HBO1 and demonstrate its mode of activity as a competitive analogue of acetyl-CoA. Inhibition of HBO1 phenocopied our genetic data and showed efficacy in a broad range of human cell lines and primary AML cells from patients. These biological, structural and chemical insights into a therapeutic target in AML will enable the clinical translation of these findings.
Collapse
Affiliation(s)
- Laura MacPherson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Juliana Anokye
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Miriam M Yeung
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Enid Y N Lam
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Yih-Chih Chan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Chen-Fang Weng
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Paul Yeh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Kathy Knezevic
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Miriam S Butler
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Annabelle Hoegl
- Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kah-Lok Chan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Marian L Burr
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Linden J Gearing
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Tracy Willson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Joy Liu
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Jarny Choi
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Yuqing Yang
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca A Bilardi
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hendrik Falk
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
| | - Nghi Nguyen
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Paul A Stupple
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Thomas S Peat
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Biomedical Program, Parkville, Victoria, Australia
| | - Ming Zhang
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
| | - Melanie de Silva
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
| | - Catalina Carrasco-Pozo
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Vicky M Avery
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Poh Sim Khoo
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Children's Cancer Institute, Kensington, New South Wales, Australia
| | - Olan Dolezal
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Biomedical Program, Parkville, Victoria, Australia
| | - Matthew L Dennis
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Biomedical Program, Parkville, Victoria, Australia
| | - Stewart Nuttall
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Biomedical Program, Parkville, Victoria, Australia
| | - Regina Surjadi
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Biomedical Program, Parkville, Victoria, Australia
| | - Janet Newman
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Biomedical Program, Parkville, Victoria, Australia
| | - Bin Ren
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Biomedical Program, Parkville, Victoria, Australia
| | - David J Leaver
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Yuxin Sun
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Jonathan B Baell
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | - Oliver Dovey
- Wellcome-MRC Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, UK
| | - George S Vassiliou
- Wellcome-MRC Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, UK
- Haematological Cancer Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, Victoria, Australia
| | - Ian P Street
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
| | - Brendon J Monahan
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Cancer Therapeutics CRC, Melbourne, Victoria, Australia
| | - Christopher J Burns
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Chunaram Choudhary
- Department of Proteomics, The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marnie E Blewitt
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anne K Voss
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Tim Thomas
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.
- Centre for Cancer Research, University of Melbourne, Melbourne, Victoria, Australia.
- Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| |
Collapse
|
263
|
|
264
|
Barbosa K, Li S, Adams PD, Deshpande AJ. The role of TP53 in acute myeloid leukemia: Challenges and opportunities. Genes Chromosomes Cancer 2019; 58:875-888. [PMID: 31393631 PMCID: PMC12042961 DOI: 10.1002/gcc.22796] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Abstract
The tumor suppressor gene TP53 is one of the most frequently mutated genes in human cancer. The central role of the TP53 protein in several fundamental processes such as cancer, aging, senescence, and DNA repair has ensured enormous attention. However, the role of TP53 in acute myeloid leukemia (AML) is enigmatic. Unlike many other human cancers, a vast majority of AMLs display no genomic TP53 alterations. There is now growing appreciation of the fact that the unaltered TP53 status of tumor cells can be exploited therapeutically. As most AMLs have an intact TP53 gene, its physiological tumor-suppressive roles could be harnessed. Therefore, the use of pharmacological activators of the TP53 pathway may provide clinical benefit in AML. Conversely, even though the frequency of TP53 mutations in AML is substantially lower than in other human cancers, TP53 mutations are associated with chemoresistance and high risk of relapse. In patients with TP53 mutations, these alterations may lead to novel, selective vulnerabilities, creating opportunities for therapeutic targeting of TP53 mutant AML. The mutational status of TP53 therefore poses challenges and opportunities in terms of advancing effective treatment strategies in AML. An increasing armamentarium of small-molecule activators of the TP53 pathway, and a growing understanding of molecular pathways triggered by mutant TP53 have accelerated efforts aimed at targeting TP53 function in AML. In combination with standard AML chemotherapy or emerging targeted therapies, pharmacological targeting of the TP53 pathway may provide therapeutic benefit in AML.
Collapse
Affiliation(s)
- Karina Barbosa
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Sha Li
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Peter D Adams
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Aniruddha J Deshpande
- Tumor Initiation and Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| |
Collapse
|
265
|
Sachs K, Sarver AL, Noble-Orcutt KE, LaRue RS, Antony ML, Chang D, Lee Y, Navis CM, Hillesheim AL, Nykaza IR, Ha NA, Hansen CJ, Karadag FK, Bergerson RJ, Verneris MR, Meredith MM, Schomaker ML, Linden MA, Myers CL, Largaespada DA, Sachs Z. Single-Cell Gene Expression Analyses Reveal Distinct Self-Renewing and Proliferating Subsets in the Leukemia Stem Cell Compartment in Acute Myeloid Leukemia. Cancer Res 2019; 80:458-470. [PMID: 31784425 DOI: 10.1158/0008-5472.can-18-2932] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 05/30/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
Standard chemotherapy for acute myeloid leukemia (AML) targets proliferative cells and efficiently induces complete remission; however, many patients relapse and die of their disease. Relapse is caused by leukemia stem cells (LSC), the cells with self-renewal capacity. Self-renewal and proliferation are separate functions in normal hematopoietic stem cells (HSC) in steady-state conditions. If these functions are also separate functions in LSCs, then antiproliferative therapies may fail to target self-renewal, allowing for relapse. We investigated whether proliferation and self-renewal are separate functions in LSCs as they often are in HSCs. Distinct transcriptional profiles within LSCs of Mll-AF9/NRASG12V murine AML were identified using single-cell RNA sequencing. Single-cell qPCR revealed that these genes were also differentially expressed in primary human LSCs and normal human HSPCs. A smaller subset of these genes was upregulated in LSCs relative to HSPCs; this subset of genes constitutes "LSC-specific" genes in human AML. To assess the differences between these profiles, we identified cell surface markers, CD69 and CD36, whose genes were differentially expressed between these profiles. In vivo mouse reconstitution assays resealed that only CD69High LSCs were capable of self-renewal and were poorly proliferative. In contrast, CD36High LSCs were unable to transplant leukemia but were highly proliferative. These data demonstrate that the transcriptional foundations of self-renewal and proliferation are distinct in LSCs as they often are in normal stem cells and suggest that therapeutic strategies that target self-renewal, in addition to proliferation, are critical to prevent relapse and improve survival in AML. SIGNIFICANCE: These findings define and functionally validate a self-renewal gene profile of leukemia stem cells at the single-cell level and demonstrate that self-renewal and proliferation are distinct in AML. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/3/458/F1.large.jpg.
Collapse
Affiliation(s)
- Karen Sachs
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,Next Generation Analytics, Palo Alto, California
| | - Aaron L Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Klara E Noble-Orcutt
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Rebecca S LaRue
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Marie Lue Antony
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Daniel Chang
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Yoonkyu Lee
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Connor M Navis
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Alexandria L Hillesheim
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Ian R Nykaza
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Ngoc A Ha
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Conner J Hansen
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Fatma K Karadag
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Rachel J Bergerson
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Michael R Verneris
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Matthew M Meredith
- Molecular Lab, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Matthew L Schomaker
- Molecular Lab, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Michael A Linden
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Chad L Myers
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Zohar Sachs
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota. .,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
266
|
RAF Kinase Inhibitor Protein in Myeloid Leukemogenesis. Int J Mol Sci 2019; 20:ijms20225756. [PMID: 31744053 PMCID: PMC6888401 DOI: 10.3390/ijms20225756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023] Open
Abstract
RAF kinase inhibitor protein (RKIP) is an essential regulator of intracellular signaling. A somatic loss of RKIP expression is a frequent event in solid human cancers, and a role of RKIP as metastasis-suppressor is widely accepted nowadays. Recently, RKIP loss has been described in acute myeloid leukemia (AML) and a series of other myeloid neoplasias (MNs). Functional in vitro and in vivo experiments revealed that RKIP is an essential player within the development of these liquid tumors; however, the respective role of RKIP seems to be complex and multi-faceted. In this review, we will summarize the current knowledge about RKIP in myeloid leukemogenesis. We will initially describe its involvement in physiologic hematopoiesis, and will then proceed to discuss its role in the development of AML and other MNs. Finally, we will discuss potential therapeutic implications arising thereof.
Collapse
|
267
|
Caiado F, Maia-Silva D, Jardim C, Schmolka N, Carvalho T, Reforço C, Faria R, Kolundzija B, Simões AE, Baubec T, Vakoc CR, da Silva MG, Manz MG, Schumacher TN, Norell H, Silva-Santos B. Lineage tracing of acute myeloid leukemia reveals the impact of hypomethylating agents on chemoresistance selection. Nat Commun 2019; 10:4986. [PMID: 31676777 PMCID: PMC6825213 DOI: 10.1038/s41467-019-12983-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy-resistant cancer recurrence is a major cause of mortality. In acute myeloid leukemia (AML), chemorefractory relapses result from the complex interplay between altered genetic, epigenetic and transcriptional states in leukemic cells. Here, we develop an experimental model system using in vitro lineage tracing coupled with exome, transcriptome and in vivo functional readouts to assess the AML population dynamics and associated molecular determinants underpinning chemoresistance development. We find that combining standard chemotherapeutic regimens with low doses of DNA methyltransferase inhibitors (DNMTi, hypomethylating drugs) prevents chemoresistant relapses. Mechanistically, DNMTi suppresses the outgrowth of a pre-determined set of chemoresistant AML clones with stemness properties, instead favoring the expansion of rarer and unfit chemosensitive clones. Importantly, we confirm the capacity of DNMTi combination to suppress stemness-dependent chemoresistance development in xenotransplantation models and primary AML patient samples. Together, these results support the potential of DNMTi combination treatment to circumvent the development of chemorefractory AML relapses. The development of post-chemotherapy resistance is a significant issue in the management of AML. Here, Caiado et al. suggest that the issue might be circumvented via upfront combination with hypomethylating agents that shape the clonal dynamics and transcriptional landscape of relapsing AML
Collapse
Affiliation(s)
- Francisco Caiado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Diogo Maia-Silva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Carolina Jardim
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Nina Schmolka
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Tânia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia Reforço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rita Faria
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Branka Kolundzija
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - André E Simões
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tuncay Baubec
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | | | | | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zürich, Switzerland
| | | | - Håkan Norell
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
268
|
Cortes JE, Dombret H, Merchant A, Tauchi T, DiRienzo CG, Sleight B, Zhang X, Leip EP, Shaik N, Bell T, Chan G, Sekeres MA. Glasdegib plus intensive/nonintensive chemotherapy in untreated acute myeloid leukemia: BRIGHT AML 1019 Phase III trials. Future Oncol 2019; 15:3531-3545. [DOI: 10.2217/fon-2019-0373] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glasdegib, an oral Hedgehog pathway inhibitor, has been associated with significantly improved survival when combined with low-dose cytarabine in patients with untreated acute myeloid leukemia (AML) who were unsuitable for intensive chemotherapy, when compared with low-dose cytarabine alone. BRIGHT AML 1019 (NCT03416179) comprises two independently powered Phase III, randomized (1:1), double-blind global trials evaluating oral glasdegib 100 mg once daily or placebo plus one of two standard chemotherapy regimens in adults with untreated AML. The intensive trial combines glasdegib/placebo with cytarabine and daunorubicin (7 + 3), while the nonintensive trial combines glasdegib/placebo with azacitidine. The primary end point of both studies is overall survival. Secondary end points include response, time to and duration of response, event-free survival, safety, patient-reported outcomes and pharmacokinetics. Trial registration number: ClinicalTrials.gov identifier: NCT03416179
Collapse
Affiliation(s)
- Jorge E Cortes
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Hervé Dombret
- Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris Institut Universitaire d`Hematologie Hopital Saint Louis, Paris, France
| | - Akil Merchant
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Tetsuzo Tauchi
- Department of Hematology, Shin-Yurigaoka General Hospital, Kawasaki, Japan
| | | | | | - Xiaoxi Zhang
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | - Eric P Leip
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | - Naveed Shaik
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | - Timothy Bell
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | - Geoffrey Chan
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | | |
Collapse
|
269
|
Long noncoding RNA MIAT promotes the progression of acute myeloid leukemia by negatively regulating miR-495. Leuk Res 2019; 87:106265. [PMID: 31698307 DOI: 10.1016/j.leukres.2019.106265] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
Abstract
Acute myeloid leukemia (AML) is a malignant myeloid hematopoietic stem and progenitor cell disease. Studies have shown that the long noncoding RNA (lncRNA) myocardial infarction associated transcript (MIAT) is abundantly expressed in multiple human solid tumors. However, the expression and role of MIAT in AML has not been explored previously. In this study, we find that MIAT is overexpressed in AML patient specimens and AML cell lines. Importantly, upregulation of MIAT is closely related with poor clinical outcome. Further investigations reveal that knockdown of MIAT inhibits the colony formation and proliferation, meanwhile, accelerates the apoptosis of AML cells in vitro. Consistently, MIAT knockdown slows AML progression in immunodeficient mice. Mechanistically, we confirm that MIAT can function as a sponge to inhibit microRNA-495 (miR-495), a tumor suppressor, in AML cells. Collectively, our results demonstrate that MIAT is involved in promoting the progression of AML, at least partly, through negative regulation of miR-495, and therefore provide a promising target for treatment of AML.
Collapse
|
270
|
Conserva MR, Redavid I, Anelli L, Zagaria A, Specchia G, Albano F. RARG Gene Dysregulation in Acute Myeloid Leukemia. Front Mol Biosci 2019; 6:114. [PMID: 31709264 PMCID: PMC6822255 DOI: 10.3389/fmolb.2019.00114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022] Open
Abstract
Retinoic acid receptor γ (RARγ) belongs to the nuclear receptor superfamily and shares 90% homology with retinoic acid receptor α (RARα) and retinoic acid receptor β (RARβ). RARA rearrangements are well-known to be involved in acute promyelocytic leukemia (APL), but RARG rearrangements can also resemble this kind of leukemia. In this review we trace the role of RARγ, considering both its physiological and oncogenic contribution; from 2011 to date, nine cases of patients harboring RARG fusions have been reported. These patients showed typical APL features, including the clinical presentation, coagulation abnormalities and morphological features of bone marrow (BM), but are not responsive to APL standard therapy. We stress the urgent need for a better comprehension of the critical role of RARG dysregulation in the leukemogenesis process, since optimum therapy strategies have not yet been established.
Collapse
Affiliation(s)
- Maria Rosa Conserva
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari, Bari, Italy
| | - Immacolata Redavid
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari, Bari, Italy
| | - Luisa Anelli
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari, Bari, Italy
| | - Antonella Zagaria
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari, Bari, Italy
| | - Giorgina Specchia
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari, Bari, Italy
| | - Francesco Albano
- Hematology Section, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari, Bari, Italy
| |
Collapse
|
271
|
Tian M, Gong W, Guo J. Long non-coding RNA SNHG1 indicates poor prognosis and facilitates disease progression in acute myeloid leukemia. Biol Open 2019; 8:bio046417. [PMID: 31615767 PMCID: PMC6826290 DOI: 10.1242/bio.046417] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/26/2019] [Indexed: 01/11/2023] Open
Abstract
The role of long non-coding RNAs (lncRNAs) in acute myeloid leukemia (AML) is becoming increasingly questioned. Previous studies have reported that the lncRNA small nucleolar RNA host gene 1 (SNHG1) is involved in multiple human malignant tumors, while its expression and role in AML is still unexplored. Here, we show that SNHG1 is highly expressed in AML specimens from non-M3 patients, as well as AML cell lines. Meanwhile, upregulation of SNHG1 is correlated with poor prognosis. Notably, SNHG1 facilitates the proliferation and inhibits the apoptosis of AML cells in vitro Consistent with these findings, knockdown of SNHG1 significantly inhibits AML progression in an immunodeficient mouse model. Mechanistically, we found that an anti-tumor microRNA-101 (miR-101) is upregulated and its target genes are downregulated in AML cells after SNHG1 knockdown. Further investigations display that SNHG1 can serve as a competing endogenous RNA to inhibit miR-101. In conclusion, our data indicate that SNHG1 plays an important role in facilitating AML progression at least in part by negatively regulating miR-101, and provides a new target for treating AML.
Collapse
Affiliation(s)
- Ming Tian
- Department of Hematology, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443000, China
| | - Wanjun Gong
- Department of Gastrointestinal Surgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443000, China
| | - Jingming Guo
- Department of Hematology, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443000, China
| |
Collapse
|
272
|
PLCB4 upregulation is associated with unfavorable prognosis in pediatric acute myeloid leukemia. Oncol Lett 2019; 18:6057-6065. [PMID: 31788080 PMCID: PMC6865073 DOI: 10.3892/ol.2019.10921] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Phospholipase C (PLC) is a membrane-associated enzyme that regulates several cellular behaviors including cell motility, growth, transformation and differentiation. PLC is involved in cancer migration, invasion and drug resistance. However, the expression status and prognostic role of PLCB4 in acute myeloid leukemia (AML) remain unclear. In the present study, the complete clinical and mRNA expression data of 285 pediatric patients with de novo AML were obtained from the Therapeutically Available Research to Generate Effective Treatments database. The association between PLCB4 expression and clinical and molecular features was explored. The expression of PLCB4 was significantly higher in patients with AML who relapsed compared with those with long-term complete remission. Patients with PLCB4 upregulation had significantly lower overall survival (OS) and event free survival (EFS) rate compared with those with low PLCB4 expression. Multivariate Cox's regression analyses demonstrated that high PLCB4 expression was an independent risk factor of adverse OS (P<0.01; HR, 2.081) and EFS (P<0.01; HR, 2.130). Following stratification analysis according to transplant status in cases of first complete remission, the patients with high expression of PLCB4 had significantly lower OS and EFS rate in the chemotherapy group, but not the stem cell transplant group. Furthermore, PLCB4-associated genes were identified using Spearman's rank correlation analysis. KEGG pathway analysis revealed that PLCB4 and its associated genes were mainly involved in three potential pathways, including the Rap1 signaling pathway. Overall, the findings of the present study suggest that increased PLCB4 expression is associated with poor clinical outcome in pediatric patients with AML, and thus may represent a potential prognostic biomarker and therapeutic target for AML.
Collapse
|
273
|
Mattes K, Vellenga E, Schepers H. Differential redox-regulation and mitochondrial dynamics in normal and leukemic hematopoietic stem cells: A potential window for leukemia therapy. Crit Rev Oncol Hematol 2019; 144:102814. [PMID: 31593878 DOI: 10.1016/j.critrevonc.2019.102814] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/12/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
The prognosis for many patients with acute myeloid leukemia (AML) is poor, mainly due to disease relapse driven by leukemia stem cells (LSCs). Recent studies have highlighted the unique metabolic properties of LSCs, which might represent opportunities for LSC-selective targeting. LSCs characteristically have low levels of reactive oxygen species (ROS), which apparently result from a combination of low mitochondrial activity and high activity of ROS-removing pathways such as autophagy. Due to this low activity, LSCs are highly dependent on mitochondrial regulatory mechanisms. These include the anti-apoptotic protein BCL-2, which also has crucial roles in regulating the mitochondrial membrane potential, and proteins involved in mitophagy. Here we review the different pathways that impact mitochondrial activity and redox-regulation, and highlight their relevance for the functionality of both HSCs and LSCs. Additionally, novel AML therapy strategies that are based on interference with those pathways, including the promising BCL-2 inhibitor Venetoclax, are summarized.
Collapse
Affiliation(s)
- Katharina Mattes
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Edo Vellenga
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hein Schepers
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
274
|
Wurm AA, Pina C. Long Non-coding RNAs as Functional and Structural Chromatin Modulators in Acute Myeloid Leukemia. Front Oncol 2019; 9:899. [PMID: 31572684 PMCID: PMC6749032 DOI: 10.3389/fonc.2019.00899] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/29/2019] [Indexed: 01/17/2023] Open
Abstract
Acute myeloid leukemia is a hematopoietic neoplasm of dismal prognosis that results from the accumulation of immature myeloid blasts in the bone marrow and the peripheral blood. It is strongly dependent on epigenetic regulation for disease onset, maintenance and in response to treatment. Epigenetic regulation refers to the multiple chemical modifications of DNA or DNA-associated proteins that alter chromatin structure and DNA accessibility in a heritable manner, without changing DNA sequence. Unlike sequence-specific transcription factors, epigenetic regulators do not necessarily bind DNA at consensus sequences, but still achieve reproducible target binding in a manner that is cell and maturation-type specific. A growing body of evidence indicates that epigenetic regulators rely, amongst other factors, on their interaction with untranslated RNA molecules for guidance to particular targets on DNA. Non (protein)-coding RNAs are the most abundant transcriptional products of the coding genome, and comprise several different classes of molecules with unique lengths, conformations and targets. Amongst these, long non-coding RNAs (lncRNAs) are species of 200 bp to >100 K bp in length, that recognize, and bind unique and largely uncharacterized DNA conformations. Some have been shown to bind epigenetic regulators, and thus constitute attractive candidates to mediate epigenetic target specificity. Herein, we postulate that lncRNAs are central players in the unique epigenetic programming of AML and review recent evidence in support of this view. We discuss the value of lncRNAs as putative diagnostic, prognostic and therapeutic targets in myeloid leukemias and indicate novel directions in this exciting research field.
Collapse
Affiliation(s)
- Alexander A Wurm
- Department of Medical Translational Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Cristina Pina
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
275
|
Shallis RM, Bewersdorf JP, Boddu PC, Zeidan AM. Hedgehog pathway inhibition as a therapeutic target in acute myeloid leukemia. Expert Rev Anticancer Ther 2019; 19:717-729. [PMID: 31422721 DOI: 10.1080/14737140.2019.1652095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: The Hedgehog (HH) pathway constitutes a collection of signaling molecules which critically influence embryogenesis. In adults, however, the HH pathway remains integral to the proliferation, maintenance, and apoptosis of adult stem cells including hematopoietic stem cells. Areas covered: We discuss the current understanding of the HH pathway as it relates to normal hematopoiesis, the pathology of acute myeloid leukemia (AML), the rationale for and data from combination therapies including HH pathway inhibitors, and ultimately the prospects that might offer promise in targeting this pathway in AML. Expert opinion: Efforts to target the HH pathway have been focused on impeding this disposition and restoring chemosensitivity to conventional myeloid neoplasm therapies. The year 2018 saw the first approval of a HH pathway inhibitor (glasdegib) for AML, though for an older population and in combination with an uncommonly-used therapy. Several other clinical trials with agents targeting modulators of HH signaling in AML and MDS are underway. Further study and understanding of the interplay between the numerous aspects of HH signaling and how it relates to the augmented survival of AML will provide a more reliable substrate for therapeutic strategies in patients with this poor-risk disease.
Collapse
Affiliation(s)
- Rory M Shallis
- Division of Hematology, Department of Medicine, Yale University School of Medicine , New Haven , CT , USA
| | - Jan Philipp Bewersdorf
- Division of Hematology, Department of Medicine, Yale University School of Medicine , New Haven , CT , USA
| | - Prajwal C Boddu
- Division of Hematology, Department of Medicine, Yale University School of Medicine , New Haven , CT , USA
| | - Amer M Zeidan
- Division of Hematology, Department of Medicine, Yale University School of Medicine , New Haven , CT , USA.,Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University , New Haven , CT , USA
| |
Collapse
|
276
|
Illangeswaran RSS, Das S, Paul DZ, Mathews V, Balasubramanian P. A personalized approach to acute myeloid leukemia therapy: current options. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:167-179. [PMID: 31447578 PMCID: PMC6684879 DOI: 10.2147/pgpm.s168267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
Therapeutic options for acute myeloid leukemia (AML) have remained unchanged for nearly the past 5 decades, with cytarabine and anthracyclines and use of hypomethylating agents for less intensive therapy. Implementation of large-scale genomic studies in the past decade has unraveled the genetic landscape and molecular etiology of AML. The approval of several novel drugs for targeted therapy, including midostaurin, enasidenib, ivosidenib, gemtuzumab–ozogamicin, and CPX351 by the US Food and Drug Administration has widened the treatment options for clinicians treating AML. This review focuses on some of these novel therapies and other promising agents under development, along with key clinical trial findings in AML.
Collapse
Affiliation(s)
| | - Saswati Das
- Department of Haematology, Christian Medical College, Vellore, India
| | | | - Vikram Mathews
- Department of Haematology, Christian Medical College, Vellore, India
| | | |
Collapse
|
277
|
Hokland P, Woll PS, Hansen MC, Bill M. The concept of leukaemic stem cells in acute myeloid leukaemia 25 years on: hitting a moving target. Br J Haematol 2019; 187:144-156. [PMID: 31372979 DOI: 10.1111/bjh.16104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The concept of leukaemic stem cells (LSCs) was experimentally suggested 25 years ago through seminal data from John Dick's group, who showed that a small fraction of cells from acute myeloid leukaemia (AML) patients were able to be adoptively transferred into immunodeficient mice. The initial estimation of the frequency was 1:250 000 leukaemic cells, clearly indicating the difficulties ahead in translating knowledge on LSCs to the clinical setting. However, the field has steadily grown in interest, expanse and importance, concomitantly with the realisation of the molecular background for AML culminating in the sequencing of hundreds of AML genomes. The literature is now ripe with contributions describing how different molecular aberrations are more or less specific for LSCs, as well as reports showing selectivity in targeting LSCs in comparison to normal haematopoietic stem and progenitor cells. However, we argue here that these important data have not yet been fully realised within the clinical setting. In this clinically focused review, we outline the difficulties in identifying and defining LSCs at the individual patient level, with special emphasis on intraclonal heterogeneity. In addition, we suggest areas of future focus in order to realise the concept as real-time benefit for AML patients.
Collapse
Affiliation(s)
- Peter Hokland
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Petter S Woll
- Department of Medicine, Huddinge, Karolinska Institute, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Marcus C Hansen
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark.,Department of Haematology, Odense University Hospital, Odense, Denmark
| | - Marie Bill
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
278
|
Depreter B, Weening KE, Vandepoele K, Essand M, De Moerloose B, Themeli M, Cloos J, Hanekamp D, Moors I, D'hont I, Denys B, Uyttebroeck A, Van Damme A, Dedeken L, Snauwaert S, Goetgeluk G, De Munter S, Kerre T, Vandekerckhove B, Lammens T, Philippé J. TARP is an immunotherapeutic target in acute myeloid leukemia expressed in the leukemic stem cell compartment. Haematologica 2019; 105:1306-1316. [PMID: 31371409 PMCID: PMC7193481 DOI: 10.3324/haematol.2019.222612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/12/2019] [Indexed: 12/26/2022] Open
Abstract
Immunotherapeutic strategies targeting the rare leukemic stem cell compartment might provide salvage to the high relapse rates currently observed in acute myeloid leukemia (AML). We applied gene expression profiling for comparison of leukemic blasts and leukemic stem cells with their normal counterparts. Here, we show that the T-cell receptor γ chain alternate reading frame protein (TARP) is over-expressed in de novo pediatric (n=13) and adult (n=17) AML sorted leukemic stem cells and blasts compared to hematopoietic stem cells and normal myeloblasts (15 healthy controls). Moreover, TARP expression was significantly associated with a fms-like tyrosine kinase receptor-3 internal tandem duplication in pediatric AML. TARP overexpression was confirmed in AML cell lines (n=9), and was found to be absent in B-cell acute lymphocytic leukemia (n=5) and chronic myeloid leukemia (n=1). Sequencing revealed that both a classical TARP transcript, as described in breast and prostate adenocarcinoma, and an AML-specific alternative TARP transcript, were present. Protein expression levels mostly matched transcript levels. TARP was shown to reside in the cytoplasmic compartment and showed sporadic endoplasmic reticulum co-localization. TARP-T-cell receptor engineered cytotoxic T-cells in vitro killed AML cell lines and patient leukemic cells co-expressing TARP and HLA-A*0201. In conclusion, TARP qualifies as a relevant target for immunotherapeutic T-cell therapy in AML.
Collapse
Affiliation(s)
- Barbara Depreter
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Karin E Weening
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Karl Vandepoele
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Magnus Essand
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Barbara De Moerloose
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Maria Themeli
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Diana Hanekamp
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ine Moors
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Inge D'hont
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Barbara Denys
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Anne Uyttebroeck
- Department of Pediatrics, University Hospital Gasthuisberg, Louvain, Belgium
| | - An Van Damme
- Department of Pediatric Hematology Oncology, University Hospital Saint-Luc, Brussels, Belgium
| | - Laurence Dedeken
- Department of Pediatric Hematology Oncology, Queen Fabiola Children's University Hospital, Brussels, Belgium
| | - Sylvia Snauwaert
- Department of Hematology, AZ Sint-Jan Hospital Bruges, Bruges, Belgium
| | - Glenn Goetgeluk
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Stijn De Munter
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tessa Kerre
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Bart Vandekerckhove
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tim Lammens
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium .,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Jan Philippé
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
279
|
Molecular mechanisms for stemness maintenance of acute myeloid leukemia stem cells. BLOOD SCIENCE 2019; 1:77-83. [PMID: 35402786 PMCID: PMC8975089 DOI: 10.1097/bs9.0000000000000020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/17/2019] [Indexed: 11/26/2022] Open
Abstract
Human acute myeloid leukemia (AML) is a fatal hematologic malignancy characterized with accumulation of myeloid blasts and differentiation arrest. The development of AML is associated with a serial of genetic and epigenetic alterations mainly occurred in hematopoietic stem and progenitor cells (HSPCs), which change HSPC state at the molecular and cellular levels and transform them into leukemia stem cells (LSCs). LSCs play critical roles in leukemia initiation, progression, and relapse, and need to be eradicated to achieve a cure in clinic. Key to successfully targeting LSCs is to fully understand the unique cellular and molecular mechanisms for maintaining their stemness. Here, we discuss LSCs in AML with a focus on identification of unique biological features of these stem cells to decipher the molecular mechanisms of LSC maintenance.
Collapse
|
280
|
Tumor heterogeneity of acute myeloid leukemia: insights from single-cell sequencing. BLOOD SCIENCE 2019; 1:73-76. [PMID: 35402804 PMCID: PMC8975104 DOI: 10.1097/bs9.0000000000000015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/11/2019] [Indexed: 11/25/2022] Open
Abstract
Individual tumors comprise genetically and epigenetically heterogeneous subclones, each of which is presumably associated with a distinct function, such as self-renewal or drug sensitivity. The dissection of such intratumoral heterogeneity is crucial to understand how tumors evolve during disease progression and under the selection of therapeutic intervention. As a paradigm of cancer intratumoral heterogeneity and clonal evolution, acute myeloid leukemia (AML) has been shown to possess complex clonal architecture based on karyotype studies, as well as deep sequencing of mixed cellular populations using next-generation sequencing (NGS) technologies. The recent development of single-cell sequencing (SCS) methods provides a powerful tool to allow analysis of genomes, transcriptomes, proteomes, and epigenomes at an individual cell level. The technologies applied in AML have broadened our understanding of AML heterogeneity and provided new insights for the development of novel therapeutic strategies. In this review, we summarize the progress in the research of AML heterogeneity using SCS technology and discuss the limitations and future direction regarding how SCS can contribute to AML prognosis and treatment.
Collapse
|
281
|
Wang X, Ghareeb WM, Zhang Y, Yu Q, Lu X, Huang Y, Huang S, Sun Y, Chi P. Hypermethylated and downregulated MEIS2 are involved in stemness properties and oxaliplatin-based chemotherapy resistance of colorectal cancer. J Cell Physiol 2019; 234:18180-18191. [PMID: 30859572 DOI: 10.1002/jcp.28451] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/26/2019] [Accepted: 01/30/2019] [Indexed: 12/16/2022]
Abstract
The resistance against oxaliplatin (L-OHP) based regimens remains a major obstacle for its efficient usage in treating metastatic colorectal cancer (mCRC). In this study, we performed weighted gene coexpression network analysis (WGCNA) to systematically screen the relevant hub genes for L-OHP resistance using the raw microarray data of 30 consecutive mCRC samples from our earlier study (GSE69657). The results were further confirmed through datasets from Gene Expression Omnibus (GEO). From L-OHP resistance module, nine genes in both the coexpression and protein-protein interaction networks were chosen as hub genes. Among these genes, Meis Homeobox 2 (MEIS2) had the highest correlation with L-OHP resistance (r = -0.443) and was deregulated in L-OHP resistant tissues compared with L-OHP sensitive tissues in both our own dataset and GSE104645 testing dataset. The receiver operating characteristic curve validated that MEIS2 had a good ability in predicting L-OHP response in both our own dataset (area under the curve [AUC] = 0.802) and GSE104645 dataset (AUC = 0.746). Then, the down expression of MEIS2 was observed in CRC tissue compared with normal tissue in 12 GEO-sourced datasets and The Cancer Genome Atlas (TCGA) and was correlated with poor event-free survival. Furthermore, analyzing methylation data from TCGA showed that MEIS2 had increased promoter hypermethylation. In addition, MEIS2 expression was significantly decreased in CRC stem cells compared with nonstem cells in two GEO datasets (GSE14773 and GSE24747). Further methylation analysis from GSE104271 demonstrated that CRC stem cells had higher MEIS2 promoter methylation levels in cg00366722 and cg00610348 sites. Gene set enrichment analysis showed that MEIS2 might be involved in the Wnt/β-catenin pathway. In the overall view, MEIS2 had increased promoter hypermethylation and was downregulated in poor L-OHP response mCRC tissues. MEIS2 might be involved in the Wnt/β-catenin pathway to maintain CRC stemness, which leads to L-OHP resistance.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Waleed M Ghareeb
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of General and Gastrointestinal Surgery, Suez Canal University, Ismailia, Egypt
| | - Yiyi Zhang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Qian Yu
- Department of Pathology, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xingrong Lu
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Ying Huang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shenghui Huang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yanwu Sun
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Pan Chi
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
282
|
Not Only Mutations Matter: Molecular Picture of Acute Myeloid Leukemia Emerging from Transcriptome Studies. JOURNAL OF ONCOLOGY 2019; 2019:7239206. [PMID: 31467542 PMCID: PMC6699387 DOI: 10.1155/2019/7239206] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/12/2019] [Indexed: 01/08/2023]
Abstract
The last two decades of genome-scale research revealed a complex molecular picture of acute myeloid leukemia (AML). On the one hand, a number of mutations were discovered and associated with AML diagnosis and prognosis; some of them were introduced into diagnostic tests. On the other hand, transcriptome studies, which preceded AML exome and genome sequencing, remained poorly translated into clinics. Nevertheless, gene expression studies significantly contributed to the elucidation of AML pathogenesis and indicated potential therapeutic directions. The power of transcriptomic approach lies in its comprehensiveness; we can observe how genome manifests its function in a particular type of cells and follow many genes in one test. Moreover, gene expression measurement can be combined with mutation detection, as high-impact mutations are often present in transcripts. This review sums up 20 years of transcriptome research devoted to AML. Gene expression profiling (GEP) revealed signatures distinctive for selected AML subtypes and uncovered the additional within-subtype heterogeneity. The results were particularly valuable in the case of AML with normal karyotype which concerns up to 50% of AML cases. With the use of GEP, new classes of the disease were identified and prognostic predictors were proposed. A plenty of genes were detected as overexpressed in AML when compared to healthy control, including KIT, BAALC, ERG, MN1, CDX2, WT1, PRAME, and HOX genes. High expression of these genes constitutes usually an unfavorable prognostic factor. Upregulation of FLT3 and NPM1 genes, independent on their mutation status, was also reported in AML and correlated with poor outcome. However, transcriptome is not limited to the protein-coding genes; other types of RNA molecules exist in a cell and regulate genome function. It was shown that microRNA (miRNA) profiles differentiated AML groups and predicted outcome not worse than protein-coding gene profiles. For example, upregulation of miR-10a, miR-10b, and miR-196b and downregulation of miR-192 were found as typical of AML with NPM1 mutation whereas overexpression of miR-155 was associated with FLT3-internal tandem duplication (FLT3-ITD). Development of high-throughput technologies and microarray replacement by next generation sequencing (RNA-seq) enabled uncovering a real variety of leukemic cell transcriptomes, reflected by gene fusions, chimeric RNAs, alternatively spliced transcripts, miRNAs, piRNAs, long noncoding RNAs (lncRNAs), and their special type, circular RNAs. Many of them can be considered as AML biomarkers and potential therapeutic targets. The relations between particular RNA puzzles and other components of leukemic cells and their microenvironment, such as exosomes, are now under investigation. Hopefully, the results of this research will shed the light on these aspects of AML pathogenesis which are still not completely understood.
Collapse
|
283
|
Bell CC, Fennell KA, Chan YC, Rambow F, Yeung MM, Vassiliadis D, Lara L, Yeh P, Martelotto LG, Rogiers A, Kremer BE, Barbash O, Mohammad HP, Johanson TM, Burr ML, Dhar A, Karpinich N, Tian L, Tyler DS, MacPherson L, Shi J, Pinnawala N, Yew Fong C, Papenfuss AT, Grimmond SM, Dawson SJ, Allan RS, Kruger RG, Vakoc CR, Goode DL, Naik SH, Gilan O, Lam EYN, Marine JC, Prinjha RK, Dawson MA. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia. Nat Commun 2019; 10:2723. [PMID: 31222014 PMCID: PMC6586637 DOI: 10.1038/s41467-019-10652-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/21/2019] [Indexed: 12/16/2022] Open
Abstract
Non-genetic drug resistance is increasingly recognised in various cancers. Molecular insights into this process are lacking and it is unknown whether stable non-genetic resistance can be overcome. Using single cell RNA-sequencing of paired drug naïve and resistant AML patient samples and cellular barcoding in a unique mouse model of non-genetic resistance, here we demonstrate that transcriptional plasticity drives stable epigenetic resistance. With a CRISPR-Cas9 screen we identify regulators of enhancer function as important modulators of the resistant cell state. We show that inhibition of Lsd1 (Kdm1a) is able to overcome stable epigenetic resistance by facilitating the binding of the pioneer factor, Pu.1 and cofactor, Irf8, to nucleate new enhancers that regulate the expression of key survival genes. This enhancer switching results in the re-distribution of transcriptional co-activators, including Brd4, and provides the opportunity to disable their activity and overcome epigenetic resistance. Together these findings highlight key principles to help counteract non-genetic drug resistance.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Bone Marrow/pathology
- CRISPR-Cas Systems/genetics
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Epigenesis, Genetic/drug effects
- Female
- Gene Expression Regulation, Leukemic/drug effects
- HEK293 Cells
- Humans
- Kaplan-Meier Estimate
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Inbred C57BL
- Sequence Analysis, RNA
- Single-Cell Analysis
- Trans-Activators/antagonists & inhibitors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription, Genetic/drug effects
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Charles C Bell
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Katie A Fennell
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Yih-Chih Chan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Florian Rambow
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Miriam M Yeung
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Dane Vassiliadis
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Luis Lara
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Paul Yeh
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | | | - Aljosja Rogiers
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Brandon E Kremer
- Epigenetics DPU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Olena Barbash
- Epigenetics DPU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Helai P Mohammad
- Epigenetics DPU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Timothy M Johanson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Marian L Burr
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Arindam Dhar
- Epigenetics DPU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | | | - Luyi Tian
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Dean S Tyler
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Laura MacPherson
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Junwei Shi
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nathan Pinnawala
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Chun Yew Fong
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Anthony T Papenfuss
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Sean M Grimmond
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Sarah-Jane Dawson
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Rhys S Allan
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Ryan G Kruger
- Epigenetics DPU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | | | - David L Goode
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Shalin H Naik
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Omer Gilan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Enid Y N Lam
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Rab K Prinjha
- Epigenetics DPU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Mark A Dawson
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
- Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
284
|
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
|
285
|
CD82 supports survival of childhood acute myeloid leukemia cells via activation of Wnt/β-catenin signaling pathway. Pediatr Res 2019; 85:1024-1031. [PMID: 30862962 DOI: 10.1038/s41390-019-0370-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/11/2019] [Accepted: 02/27/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND Stem cell marker CD82 plays a vital role in the oncogenesis and progression of acute myelogenous leukemia (AML), especially in sharing properties of leukemia stem cells (LSCs). The Wnt/β-catenin pathway is required for the development of LSCs in AML. The present study aimed to validate whether CD82 supports the survival of LSCs in pediatric AML via activation of Wnt/β-catenin signaling pathway. METHODS CD82 expression and its correlation with molecules downstream of Wnt/β-catenin pathway in samples from pediatric AML patients were analyzed. Forced or downregulated expression of CD82 in AML cells was evaluated for the effects of CD82 on cell proliferation, cycle regulation, apoptosis, and adriamycin chemoresistance and to validate the underlying mechanism. RESULT Aberrant expression of CD82 in pediatric AML patients was found. CD82 messenger RNA expression correlated positively with downstream molecules of Wnt/β-catenin pathway in AML children. Knockdown of CD82 induced apoptosis, suppressed growth, and decreased adriamycin chemoresistance in AML cells. CD82 accelerated β-catenin nuclear location and then stimulated the expression of downstream molecules of Wnt/β-catenin pathway. CONCLUSION CD82 regulates the proliferation and chemotherapy resistance of AML cells via activation of the Wnt/β-catenin pathway, which suggest that the CD82 may be a potential therapeutic target in AML children.
Collapse
|
286
|
Li J, Lu L, Zhang YH, Xu Y, Liu M, Feng K, Chen L, Kong X, Huang T, Cai YD. Identification of leukemia stem cell expression signatures through Monte Carlo feature selection strategy and support vector machine. Cancer Gene Ther 2019; 27:56-69. [PMID: 31138902 DOI: 10.1038/s41417-019-0105-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/28/2019] [Accepted: 05/04/2019] [Indexed: 01/09/2023]
Abstract
Acute myeloid leukemia (AML) is a type of blood cancer characterized by the rapid growth of immature white blood cells from the bone marrow. Therapy resistance resulting from the persistence of leukemia stem cells (LSCs) are found in numerous patients. Comparative transcriptome studies have been previously conducted to analyze differentially expressed genes between LSC+ and LSC- cells. However, these studies mainly focused on a limited number of genes with the most obvious expression differences between the two cell types. We developed a computational approach incorporating several machine learning algorithms, including Monte Carlo feature selection (MCFS), incremental feature selection (IFS), support vector machine (SVM), Repeated Incremental Pruning to Produce Error Reduction (RIPPER), to identify gene expression features specific to LSCs. One thousand 0ne hudred fifty-nine features (genes) were first identified, which can be used to build the optimal SVM classifier for distinguishing LSC+ and LSC- cells. Among these 1159 genes, the top 17 genes were identified as LSC-specific biomarkers. In addition, six classification rules were produced by RIPPER algorithm. The subsequent literature review on these features/genes and the classification rules and functional enrichment analyses of the 1159 features/genes confirmed the relevance of extracted genes and rules to the characteristics of LSCs.
Collapse
Affiliation(s)
- JiaRui Li
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China.,School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Yu-Hang Zhang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China
| | - YaoChen Xu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China
| | - Min Liu
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, P. R. China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic, Guangzhou, 510507, P. R. China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, P. R. China.,Shanghai Key Laboratory of PMMP, East China Normal University, Shanghai, 200241, P. R. China
| | - XiangYin Kong
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China.
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P. R. China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
287
|
Annageldiyev C, Gowda K, Patel T, Bhattacharya P, Tan SF, Iyer S, Desai D, Dovat S, Feith DJ, Loughran TP, Amin S, Claxton D, Sharma A. The novel Isatin analog KS99 targets stemness markers in acute myeloid leukemia. Haematologica 2019; 105:687-696. [PMID: 31123028 PMCID: PMC7049373 DOI: 10.3324/haematol.2018.212886] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 05/22/2019] [Indexed: 01/01/2023] Open
Abstract
Leukemic stem cells are multipotent, self-renewing, highly proliferative cells that can withstand drug treatments. Although currently available treatments potentially destroy blast cells, they fail to eradicate leukemic progenitor cells completely. Aldehyde dehydrogenase and STAT3 are frequently up-regulated in pre-leukemic stem cells as well as in acute myeloid leukemia (AML) expressing the CD34+CD38− phenotype. The Isatin analog, KS99 has shown anticancer activity against multiple myeloma which may, in part, be mediated by inhibition of Bruton’s tyrosine kinase activation. Here we demonstrate that KS99 selectively targets leukemic stem cells with high aldehyde dehydrogenase activity and inhibits phosphorylation of STAT3. KS99 targeted cells co-expressing CD34, CD38, CD123, TIM-3, or CD96 immunophenotypes in AML, alone or in combination with the standard therapeutic agent cytarabine. AML with myelodysplastic-related changes was more sensitive than de novo AML with or without NPM1 mutation. KS99 treatment reduced the clonogenicity of primary human AML cells as compared to normal cord blood mononuclear cells. Downregulation of phosphorylated Bruton’s tyrosine kinase, STAT3, and aldehyde dehydrogenase was observed, suggesting interaction with KS99 as predicted through docking. KS99 with or without cytarabine showed in vivo preclinical efficacy in human and mouse AML animal models and prolonged survival. KS99 was well tolerated with overall negligible adverse effects. In conclusion, KS99 inhibits aldehyde dehydrogenase and STAT3 activities and causes cell death of leukemic stem cells, but not normal hematopoietic stem and progenitor cells.
Collapse
Affiliation(s)
- Charyguly Annageldiyev
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Krishne Gowda
- Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Trupti Patel
- Department of Integrative Biotechnology, SBST, VIT Vellore, Tamilnadu, India
| | | | - Su-Fern Tan
- Department of Medicine, Division of Hematology and Oncology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Soumya Iyer
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Dhimant Desai
- Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Sinisa Dovat
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - David J Feith
- Department of Medicine, Division of Hematology and Oncology, University of Virginia School of Medicine, Charlottesville, VA, USA.,University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Thomas P Loughran
- Department of Medicine, Division of Hematology and Oncology, University of Virginia School of Medicine, Charlottesville, VA, USA.,University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Shantu Amin
- Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - David Claxton
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Arati Sharma
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, USA .,Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
288
|
Shallis RM, Wang R, Davidoff A, Ma X, Zeidan AM. Epidemiology of acute myeloid leukemia: Recent progress and enduring challenges. Blood Rev 2019; 36:70-87. [PMID: 31101526 DOI: 10.1016/j.blre.2019.04.005] [Citation(s) in RCA: 516] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 04/06/2019] [Accepted: 04/26/2019] [Indexed: 01/08/2023]
Abstract
Acute myeloid leukemia (AML) is a malignant disorder of the bone marrow which is characterized by the clonal expansion and differentiation arrest of myeloid progenitor cells. The age-adjusted incidence of AML is 4.3 per 100,000 annually in the United States (US). Incidence increases with age with a median age at diagnosis of 68 years in the US. The etiology of AML is heterogeneous. In some patients, prior exposure to therapeutic, occupational or environmental DNA-damaging agents is implicated, but most cases of AML remain without a clear etiology. AML is the most common form of acute leukemia in adults and has the shortest survival (5-year survival = 24%). Curative therapies, including intensive chemotherapy and allogeneic stem cell transplantation, are generally applicable to a minority of patients who are younger and fit, while most older individuals exhibit poor prognosis and survival. Differences in patient outcomes are influenced by disease characteristics, access to care including active therapies and supportive care, and other factors. After many years without therapeutic advances, several new therapies have been approved and are expected to impact patient outcomes, especially for older patients and those with refractory disease.
Collapse
Affiliation(s)
- Rory M Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
| | - Rong Wang
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, USA; Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, USA
| | - Amy Davidoff
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, USA; Department of Health Policy and Management, School of Public Health, Yale University, New Haven, USA
| | - Xiaomei Ma
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, USA; Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA; Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, USA.
| |
Collapse
|
289
|
Bohl SR, Bullinger L, Rücker FG. New Targeted Agents in Acute Myeloid Leukemia: New Hope on the Rise. Int J Mol Sci 2019; 20:E1983. [PMID: 31018543 PMCID: PMC6515298 DOI: 10.3390/ijms20081983] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 02/07/2023] Open
Abstract
The therapeutic approach for acute myeloid leukemia (AML) remains challenging, since over the last four decades a stagnation in standard cytotoxic treatment has been observed. But within recent years, remarkable advances in the understanding of the molecular heterogeneity and complexity of this disease have led to the identification of novel therapeutic targets. In the last two years, seven new targeted agents (midostaurin, gilteritinib, enasidenib, ivosidenib, glasdegib, venetoclax and gemtuzumab ozogamicin) have received US Food and Drug Administration (FDA) approval for the treatment of AML. These drugs did not just prove to have a clinical benefit as single agents but have especially improved AML patient outcomes if they are combined with conventional therapy. In this review, we will focus on currently approved and promising upcoming agents and we will discuss controversial aspects and limitations of targeted treatment strategies.
Collapse
Affiliation(s)
- Stephan R Bohl
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany.
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumorimmunology, Charité University Medicine, 13353 Berlin, Germany.
| | - Frank G Rücker
- Department of Internal Medicine III, University Hospital Ulm, 89081 Ulm, Germany.
| |
Collapse
|
290
|
Xu J, Nuno K, Litzenburger UM, Qi Y, Corces MR, Majeti R, Chang HY. Single-cell lineage tracing by endogenous mutations enriched in transposase accessible mitochondrial DNA. eLife 2019; 8:45105. [PMID: 30958261 PMCID: PMC6469926 DOI: 10.7554/elife.45105] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/07/2019] [Indexed: 12/22/2022] Open
Abstract
Simultaneous measurement of cell lineage and cell fates is a longstanding goal in biomedicine. Here we describe EMBLEM, a strategy to track cell lineage using endogenous mitochondrial DNA variants in ATAC-seq data. We show that somatic mutations in mitochondrial DNA can reconstruct cell lineage relationships at single cell resolution with high sensitivity and specificity. Using EMBLEM, we define the genetic and epigenomic clonal evolution of hematopoietic stem cells and their progenies in patients with acute myeloid leukemia. EMBLEM extends lineage tracing to any eukaryotic organism without genetic engineering.
Collapse
Affiliation(s)
- Jin Xu
- Center for Personal Dynamic Regulomes, Stanford, United States.,Department of Dermatology, Stanford University School of Medicine, Stanford, United States.,Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Kevin Nuno
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, United States.,Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, United States
| | - Ulrike M Litzenburger
- Center for Personal Dynamic Regulomes, Stanford, United States.,Department of Dermatology, Stanford University School of Medicine, Stanford, United States.,Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Yanyan Qi
- Center for Personal Dynamic Regulomes, Stanford, United States.,Department of Dermatology, Stanford University School of Medicine, Stanford, United States.,Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - M Ryan Corces
- Center for Personal Dynamic Regulomes, Stanford, United States.,Department of Dermatology, Stanford University School of Medicine, Stanford, United States.,Department of Genetics, Stanford University School of Medicine, Stanford, United States
| | - Ravindra Majeti
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, United States.,Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, United States
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford, United States.,Department of Dermatology, Stanford University School of Medicine, Stanford, United States.,Department of Genetics, Stanford University School of Medicine, Stanford, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, United States
| |
Collapse
|
291
|
Qin J, Bao H, Li H. Correlation of long non-coding RNA taurine-upregulated gene 1 with disease conditions and prognosis, as well as its effect on cell activities in acute myeloid leukemia. Cancer Biomark 2019; 23:569-577. [PMID: 30452403 DOI: 10.3233/cbm-181834] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE This study aimed to investigate the correlation of long non-coding RNA taurine-upregulated gene 1 (lncRNA TUG1) with clinicopathological characteristics and prognosis in acute myeloid leukemia (AML) patients, as well as its function in cell proliferation and apoptosis. METHODS Two hundred and thirty six de novo AML patients were consecutively enrolled and then underwent conventional induction chemotherapy. Bone marrow samples were obtained from all AML patients and controls. Quantitative polymerase chain reaction assay was performed to detect lncRNA TUG1 expression. KG-1 cells were transfected by TUG1 inhibitor (TUG1 (-)) and blank inhibitor (NC (-)) plasmids. Cell proliferation and apoptosis were evaluated by CCK8 and AV/PI assays, and apoptotic markers expressions were detected by Western blot assay. RESULTS LncRNA TUG1 expression was higher in AML patients compared to controls, and it was positively correlated with white blood cell counts as well as poor risk stratification. Additionally, elevated lncRNA TUG1 expression was observed in non-complete remission (non-CR) patients compared to CR patients, and it was correlated with shorter event-free survival and overall survival in AML patients. In the in vitro experiments, lncRNA TUG1 expression was upregulated in AML cell lines compared to control cells, and cell proliferation ability was reduced, but cell apoptosis rate was promoted in TUG1 (-) group compared to NC (-) group at 72 hours after transfection in KG-1 cells. CONCLUSIONS LncRNA TUG1 predicts advanced disease conditions and poor prognosis in AML patients, and its knockout decreases proliferation and increases apoptosis of AML cells.
Collapse
|
292
|
Azevedo PL, Oliveira NCA, Corrêa S, Castelo-Branco MTL, Abdelhay E, Binato R. Canonical WNT Signaling Pathway is Altered in Mesenchymal Stromal Cells From Acute Myeloid Leukemia Patients And Is Implicated in BMP4 Down-Regulation. Transl Oncol 2019; 12:614-625. [PMID: 30703678 PMCID: PMC6350721 DOI: 10.1016/j.tranon.2019.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/02/2019] [Accepted: 01/02/2019] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cells (hMSCs) are key components of the bone marrow microenvironment (BMM). A molecular signature in hMSCs from Acute myeloid leukemia patients (hMSC-AML) has been proposed where BMP4 is decreased and could be regulated by WNT signaling pathway. Therefore, the aim of this work was to verify whether the WNT signaling pathway can regulate the BMP4 gene in hMSCs. The results showed differentially expressed genes in the WNT canonical pathway between hMSC-AML and hMSCs from healthy donors and a real-time quantitative assay corroborated with these findings. Moreover, the main WNT canonical pathway regulators were decreased in hMSC-AML, such as LEF-1, β-catenin and the β-catenin/TCF-LEF regulatory complex in the nucleus. This result, together with functional assays, suggests that the induction of BMP4 expression by the WNT signaling pathway is decreased in hMSC-AML. Overall, the WNT canonical pathway is able to regulate the BMP4 gene in hMSC-AML and its reduced activation could also lead to the lower expression of BMP4 in hMSC-AML. Due to the important role of the BMM, changes in BMP4 expression through the WNT canonical pathway may be a potential mechanism of leukemogenesis.
Collapse
Affiliation(s)
- Pedro L Azevedo
- Stem Cell Laboratory, Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Nathalia C A Oliveira
- Stem Cell Laboratory, Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Stephany Corrêa
- Stem Cell Laboratory, Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Morgana T L Castelo-Branco
- Institute of Biomedical Sciences and Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Eliana Abdelhay
- Stem Cell Laboratory, Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Renata Binato
- Stem Cell Laboratory, Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
293
|
Daga S, Rosenberger A, Quehenberger F, Krisper N, Prietl B, Reinisch A, Zebisch A, Sill H, Wölfler A. High GPR56 surface expression correlates with a leukemic stem cell gene signature in CD34-positive AML. Cancer Med 2019; 8:1771-1778. [PMID: 30848055 PMCID: PMC6488118 DOI: 10.1002/cam4.2053] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/24/2019] [Accepted: 02/04/2019] [Indexed: 12/11/2022] Open
Abstract
Acute myeloid leukemia (AML) is driven by a minor fraction of leukemic stem cells (LSCs) whose persistence is considered being the primary cause of disease relapse. A detailed characterization of the surface immunophenotype of LSCs to discriminate them from bulk leukemic blasts may enable successful targeting of this population thereby improving patient outcomes in AML. To identify surface markers, which may reflect LSC activity at diagnosis, we performed a detailed analysis of 16 putative LSC markers in CD34/38 leukemic subcompartments of 150 diagnostic AML samples using multicolor flow cytometry. The most promising markers were then selected to determine a possible correlation of their expression with a recently published LSC gene signature. We found GPR56 and CLL-1 to be the most prominently differently expressed surface markers in AML subcompartments. While GPR56 was highest expressed within the LSC-enriched CD34+ 38- subcompartment as compared to CD34+ 38+ and CD34- leukemic bulk cells, CLL-1 expression was lowest in CD34+ 38- leukemic cells and increased in CD34+ 38+ and CD34- blasts. Furthermore, high GPR56 surface expression in CD34+ 38- leukemic cells correlated with a recently published LSC gene expression signature and was associated with decreased overall survival in patients receiving intensive chemotherapy. In contrast, CLL-1 expression correlated inversely with the LSC gene signature and was not informative on outcome. Our data strongly support GPR56 as a promising clinically relevant marker for identifying leukemic cells with LSC activity at diagnosis in CD34-positive AML.
Collapse
Affiliation(s)
- Shruti Daga
- Division of Hematology, Medical University of Graz, Graz, Austria.,CBmed Center of Biomarker Research in Medicine, Graz, Austria
| | - Angelika Rosenberger
- Division of Hematology, Medical University of Graz, Graz, Austria.,CBmed Center of Biomarker Research in Medicine, Graz, Austria
| | - Franz Quehenberger
- Institute of Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Nina Krisper
- CBmed Center of Biomarker Research in Medicine, Graz, Austria
| | - Barbara Prietl
- CBmed Center of Biomarker Research in Medicine, Graz, Austria.,Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Andreas Reinisch
- Division of Hematology, Medical University of Graz, Graz, Austria
| | - Armin Zebisch
- Division of Hematology, Medical University of Graz, Graz, Austria
| | - Heinz Sill
- Division of Hematology, Medical University of Graz, Graz, Austria
| | - Albert Wölfler
- Division of Hematology, Medical University of Graz, Graz, Austria.,CBmed Center of Biomarker Research in Medicine, Graz, Austria
| |
Collapse
|
294
|
Prochazka KT, Pregartner G, Rücker FG, Heitzer E, Pabst G, Wölfler A, Zebisch A, Berghold A, Döhner K, Sill H. Clinical implications of subclonal TP53 mutations in acute myeloid leukemia. Haematologica 2019; 104:516-523. [PMID: 30309854 PMCID: PMC6395341 DOI: 10.3324/haematol.2018.205013] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023] Open
Abstract
The role of subclonal TP53 mutations, defined by a variant allele frequency of <20%, has not been addressed in acute myeloid leukemia yet. We, therefore, analyzed their prognostic value in a cohort of 1,537 patients with newly diagnosed disease, prospectively treated within three trials of the "German-Austrian Acute Myeloid Leukemia Study Group". Mutational analysis was performed by targeted deep sequencing and patients with TP53 mutations were categorized by their variant allele frequency into groups with frequencies >40%, 20%-40% and <20%. A total of 108 TP53 mutations were found in 98 patients (6.4%). Among these, 61 patients had variant allele frequencies >40%, 19 had variant allele frequencies between 20%-40% and 18 had frequencies <20%. Compared to specimens with clonal TP53 mutations, those with subclonal ones showed significantly fewer complex karyotypes and chromosomal losses. In either TP53-mutated group, patients experienced significantly fewer complete responses (P<0.001) and had worse overall and event-free survival rates (P<0.0001). In Cox regression analyses adjusting for age, white blood cell count, cytogenetic risk and type of acute myeloid leukemia, the adverse prognostic effect of TP53 mutations remained significant for all TP53-mutated subgroups. These data suggest that subclonal TP53 mutations are a novel prognostic parameter in acute myeloid leukemia and emphasize the usefulness of next-generation sequencing technologies for risk stratification in this disorder. The study was registered at ClinicalTrials.gov with number NCT00146120.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Alleles
- Chromosome Aberrations
- Clonal Evolution/genetics
- Female
- Gene Frequency
- Genes, p53
- Humans
- Kaplan-Meier Estimate
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Mutation
- Prognosis
- Young Adult
Collapse
Affiliation(s)
| | - Gudrun Pregartner
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Austria
| | - Frank G Rücker
- Department of Internal Medicine III, University Hospital of Ulm, Germany
| | - Ellen Heitzer
- Institute of Human Genetics, Medical University of Graz, Austria
| | - Gabriel Pabst
- Division of Hematology, Medical University of Graz, Austria
| | - Albert Wölfler
- Division of Hematology, Medical University of Graz, Austria
| | - Armin Zebisch
- Division of Hematology, Medical University of Graz, Austria
| | - Andrea Berghold
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Austria
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Germany
| | - Heinz Sill
- Division of Hematology, Medical University of Graz, Austria
| |
Collapse
|
295
|
NCAM1 (CD56) promotes leukemogenesis and confers drug resistance in AML. Blood 2019; 133:2305-2319. [PMID: 30814062 DOI: 10.1182/blood-2018-12-889725] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023] Open
Abstract
Neural cell adhesion molecule 1 (NCAM1; CD56) is expressed in up to 20% of acute myeloid leukemia (AML) patients. NCAM1 is widely used as a marker of minimal residual disease; however, the biological function of NCAM1 in AML remains elusive. In this study, we investigated the impact of NCAM1 expression on leukemogenesis, drug resistance, and its role as a biomarker to guide therapy. Beside t(8;21) leukemia, NCAM1 expression was found in most molecular AML subgroups at highly heterogeneous expression levels. Using complementary genetic strategies, we demonstrated an essential role of NCAM1 in the regulation of cell survival and stress resistance. Perturbation of NCAM1 induced cell death or differentiation and sensitized leukemic blasts toward genotoxic agents in vitro and in vivo. Furthermore, Ncam1 was highly expressed in leukemic progenitor cells in a murine leukemia model, and genetic depletion of Ncam1 prolonged disease latency and significantly reduced leukemia-initiating cells upon serial transplantation. To further analyze the mechanism of the NCAM1-associated phenotype, we performed phosphoproteomics and transcriptomics in different AML cell lines. NCAM1 expression strongly associated with constitutive activation of the MAPK-signaling pathway, regulation of apoptosis, or glycolysis. Pharmacological inhibition of MEK1/2 specifically inhibited proliferation and sensitized NCAM1+ AML cells to chemotherapy. In summary, our data demonstrate that aberrant expression of NCAM1 is involved in the maintenance of leukemic stem cells and confers stress resistance, likely due to activation of the MAPK pathway. Targeting MEK1/2 sensitizes AML blasts to genotoxic agents, indicating a role for NCAM1 as a biomarker to guide AML treatment.
Collapse
|
296
|
Wang Y, Richter L, Becker M, Amador C, Hyde RK. IL1RL1 is dynamically expressed on Cbfb-MYH11 + leukemia stem cells and promotes cell survival. Sci Rep 2019; 9:1729. [PMID: 30742053 PMCID: PMC6370767 DOI: 10.1038/s41598-018-38408-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/21/2018] [Indexed: 11/25/2022] Open
Abstract
Acute myeloid leukemia (AML) is often characterized by the presence of specific, recurrent chromosomal abnormalities. One of the most common aberrations, inversion of chromosome 16 [inv(16)], generates the fusion oncogene CBFB-MYH11. Previously, we used a mouse knock-in model to show that Cbfb-MYH11 induces changes in gene expression and results in the accumulation of abnormal myeloid cells, a subset of which are enriched for leukemia stem cell (LSC) activity. One gene upregulated by Cbfb-MYH11 encodes the cytokine receptor IL1RL1 (ST2). IL1RL1 and its ligand IL-33 are known regulators of mature myeloid cells, but their roles in AML are not known. Here, we use Cbfb-MYH11 knock-in mice to show that IL1RL1 is expressed by cell populations with high LSC activity, and that the cell surface expression of IL1RL1 is dynamic, implying that the expression of IL1RL1 is not restricted to a specific stage of differentiation. We also show that treatment with IL-33 increased serial replating ability and expression of pro-survival proteins in vitro. Finally, we show that IL1RL1+ cells can survive chemotherapy better than IL1RL1− cells in vivo. Collectively, our results indicate that IL1RL1 is dynamically expressed in Cbfb-MYH11+ leukemia cells and promotes their survival.
Collapse
Affiliation(s)
- Yiqian Wang
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Lisa Richter
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michelle Becker
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Catalina Amador
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - R Katherine Hyde
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
297
|
Data mining for mutation-specific targets in acute myeloid leukemia. Leukemia 2019; 33:826-843. [PMID: 30728456 DOI: 10.1038/s41375-019-0387-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/06/2018] [Accepted: 10/24/2018] [Indexed: 01/08/2023]
Abstract
Three mutation-specific targeted therapies have recently been approved by the FDA for the treatment of acute myeloid leukemia (AML): midostaurin for FLT3 mutations, enasidenib for relapsed or refractory cases with IDH2 mutations, and ivosidenib for cases with an IDH1 mutation. Together, these agents offer a mutation-directed treatment approach for up to 45% of de novo adult AML cases, a welcome deluge after a prolonged drought. At the same time, a number of computational tools have recently been developed that promise to further accelerate progress in mutation-specific therapy for AML and other cancers. Technical advances together with comprehensively annotated AML tissue banks have resulted in the availability of large and complex data sets for exploration by the end-user, including (i) microarray gene expression, (ii) exome sequencing, (iii) deep sequencing data of sub-clone heterogeneity, (iv) RNA sequencing of gene expression (bulk and single cell), (v) DNA methylation and chromatin, (vi) and germline quantitative trait loci. Yet few clinicians or experimental hematologists have the time or the training to access or analyze these repositories. This review summarizes the data sets and bioinformatic tools currently available to further the discovery of mutation-specific targets with an emphasis on web-based applications that are open, accessible, user-friendly, and do not require coding experience to navigate. We show examples of how available data can be mined to identify potential targets using synthetic lethality, drug repurposing, epigenetic sub-grouping, and proteomic networks while also highlighting strengths and limitations and the need for superior models for validation.
Collapse
|
298
|
Yassin M, Aqaqe N, Yassin AA, van Galen P, Kugler E, Bernstein BE, Koren-Michowitz M, Canaani J, Nagler A, Lechman ER, Dick JE, Wienholds E, Izraeli S, Milyavsky M. A novel method for detecting the cellular stemness state in normal and leukemic human hematopoietic cells can predict disease outcome and drug sensitivity. Leukemia 2019; 33:2061-2077. [PMID: 30705411 DOI: 10.1038/s41375-019-0386-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/02/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023]
Abstract
Acute leukemia is an aggressive blood malignancy with low survival rates. A high expression of stem-like programs in leukemias predicts poor prognosis and is assumed to act in an aberrant fashion in the phenotypically heterogeneous leukemia stem cell (LSC) population. A lack of suitable genome engineering tools that can isolate LSCs based on their stemness precludes their comprehensive examination and full characterization. We hypothesized that tagging endogenous stemness-regulatory regions could generate a genome reporter for the putative leukemia stemness-state. Our analysis revealed that the ERG + 85 enhancer region can serve as a marker for stemness-state and a fluorescent lentiviral reporter was developed that can accurately recapitulate the endogenous activity. Using our novel reporter, we revealed cellular heterogeneity in several leukemia cell lines and patient-derived samples. Alterations in reporter activity were associated with transcriptomic and functional changes that were closely related to the hematopoietic stem cell (HSC) identity. Notably, the differentiation potential was skewed towards the erythro-megakaryocytic lineage. Moreover, an ERG + 85High fraction of AML cells could regenerate the original cellular heterogeneity and was enriched for LSCs. RNA-seq analysis coupled with in silico drug-screen analysis identified 4HPR as an effective inhibitor of ERG + 85High leukemia growth. We propose that further utilization of our novel molecular tool will identify crucial determinants of LSCs, thus providing a rationale for their therapeutic targeting.
Collapse
Affiliation(s)
- Muhammad Yassin
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Nasma Aqaqe
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Abed Alkader Yassin
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Peter van Galen
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - 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, Chaim Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Bradley E Bernstein
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, 02114, USA
| | | | - Jonathan Canaani
- Hematology Division, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Erno Wienholds
- Princess Margaret Cancer Centre, University Health Network and Department of Molecular Genetics, University of Toronto, Toronto, ON, 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, Chaim Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Department of Molecular Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Michael Milyavsky
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel.
| |
Collapse
|
299
|
Current Outlook on Autophagy in Human Leukemia: Foe in Cancer Stem Cells and Drug Resistance, Friend in New Therapeutic Interventions. Int J Mol Sci 2019; 20:ijms20030461. [PMID: 30678185 PMCID: PMC6387281 DOI: 10.3390/ijms20030461] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 01/07/2023] Open
Abstract
Autophagy is an evolutionarily conserved cellular recycling process in cell homeostasis and stress adaptation. It confers protection and promotes survival in response to metabolic/environmental stress, and is upregulated in response to nutrient deprivation, hypoxia, and chemotherapies. Autophagy is also known to sustain malignant cell growth and contributes to cancer stem cell survival when challenged by cytotoxic and/or targeted therapies, a potential mechanism of disease persistence and drug resistance that has gathered momentum. However, different types of human leukemia utilize autophagy in complex, context-specific manners, and the molecular and cellular mechanisms underlying this process involve multiple protein networks that will be discussed in this review. There is mounting preclinical evidence that targeting autophagy can enhance the efficacy of cancer therapies. Chloroquine and other lysosomal inhibitors have spurred initiation of clinical trials and demonstrated that inhibition of autophagy restores chemosensitivity of anticancer drugs, but with limited autophagy-dependent effects. Intriguingly, several autophagy-specific inhibitors, with better therapeutic indexes and lower toxicity, have been developed. Promising preclinical studies with novel combination approaches as well as potential challenges to effectively eradicate drug-resistant cells, particularly cancer stem cells, in human leukemia are also detailed in this review.
Collapse
|
300
|
Gruszka AM, Valli D, Restelli C, Alcalay M. Adhesion Deregulation in Acute Myeloid Leukaemia. Cells 2019; 8:E66. [PMID: 30658474 PMCID: PMC6356639 DOI: 10.3390/cells8010066] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Cell adhesion is a process through which cells interact with and attach to neighboring cells or matrix using specialized surface cell adhesion molecules (AMs). Adhesion plays an important role in normal haematopoiesis and in acute myeloid leukaemia (AML). AML blasts express many of the AMs identified on normal haematopoietic precursors. Differential expression of AMs between normal haematopoietic cells and leukaemic blasts has been documented to a variable extent, likely reflecting the heterogeneity of the disease. AMs govern a variety of processes within the bone marrow (BM), such as migration, homing, and quiescence. AML blasts home to the BM, as the AM-mediated interaction with the niche protects them from chemotherapeutic agents. On the contrary, they detach from the niches and move from the BM into the peripheral blood to colonize other sites, i.e., the spleen and liver, possibly in a process that is reminiscent of epithelial-to-mesenchymal-transition in metastatic solid cancers. The expression of AMs has a prognostic impact and there are ongoing efforts to therapeutically target adhesion in the fight against leukaemia.
Collapse
Affiliation(s)
- Alicja M Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Debora Valli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Cecilia Restelli
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
| | - Myriam Alcalay
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Via Adamello 16, 20 139 Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20 122 Milan, Italy.
| |
Collapse
|