1
|
Wang YQ, Ren Y, Gale RP, Niu LT, Huang XJ. Sphingosine-1 phosphate receptor 1 (S1PR1) expression maintains stemness of acute myeloid leukemia stem cells. Cancer Lett 2024; 600:217158. [PMID: 39111385 DOI: 10.1016/j.canlet.2024.217158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/16/2024] [Revised: 07/24/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Acute myeloid leukemia (AML) arises from leukemia stem cells (LSCs) and is maintained by cells which have acquired features of stemness. We compared transcription profiles of AML cells with/without stem cell features defined as in vitro clonogenicity and serial engraftment in immune-deficient mice xenograft model. We used multi-parameter flow cytometry (MPFC) to separate CD34+ bone marrow-derived leukemia cells into sphingosine-1 phosphate receptor 1 (S1PR1)+ and S1PR1- fractions. Cells in the S1PR1+ fraction demonstrated significantly higher clonogenicity and higher engraftment potential compared with those in the S1PR1- fraction. In contrast, CD34+ bone marrow cells from normal samples showed reduced clonogenicity in the S1PR1+ fraction compared with the S1PR1- fraction. Inhibition of S1PR1 expression in an AML cell line reduced the colony-forming potential of KG1 cells. Transcriptomic analyses and rescue experiments indicated PI3K/AKT pathway and MYBL2 are downstream mediators of S1PR1-associated stemness. These findings implicate S1PR1 as a functional biomarker of LSCs and suggest its potential as a therapeutic target in AML treatment.
Collapse
Affiliation(s)
- Yu-Qing Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yue Ren
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Robert Peter Gale
- Centre for Hematology Research, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Li-Ting Niu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
2
|
Bordeleau ME, Audemard É, Métois A, Theret L, Lisi V, Farah A, Spinella JF, Chagraoui J, Moujaber O, Aubert L, Khakipoor B, Mallinger L, Boivin I, Mayotte N, Hajmirza A, Bonneil É, Béliveau F, Pfammatter S, Feghaly A, Boucher G, Gendron P, Thibault P, Barabé F, Lemieux S, Richard-Carpentier G, Hébert J, Lavallée VP, Roux PP, Sauvageau G. Immunotherapeutic targeting of surfaceome heterogeneity in AML. Cell Rep 2024; 43:114260. [PMID: 38838225 DOI: 10.1016/j.celrep.2024.114260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/05/2023] [Revised: 01/29/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
Immunotherapy remains underexploited in acute myeloid leukemia (AML) compared to other hematological malignancies. Currently, gemtuzumab ozogamicin is the only therapeutic antibody approved for this disease. Here, to identify potential targets for immunotherapeutic intervention, we analyze the surface proteome of 100 genetically diverse primary human AML specimens for the identification of cell surface proteins and conduct single-cell transcriptome analyses on a subset of these specimens to assess antigen expression at the sub-population level. Through this comprehensive effort, we successfully identify numerous antigens and markers preferentially expressed by primitive AML cells. Many identified antigens are targeted by therapeutic antibodies currently under clinical evaluation for various cancer types, highlighting the potential therapeutic value of the approach. Importantly, this initiative uncovers AML heterogeneity at the surfaceome level, identifies several antigens and potential primitive cell markers characterizing AML subgroups, and positions immunotherapy as a promising approach to target AML subgroup specificities.
Collapse
Affiliation(s)
- Marie-Eve Bordeleau
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| | - Éric Audemard
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Arnaud Métois
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Louis Theret
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Véronique Lisi
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Azer Farah
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Jean-François Spinella
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Jalila Chagraoui
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Ossama Moujaber
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Léo Aubert
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Banafsheh Khakipoor
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Laure Mallinger
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Isabel Boivin
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Nadine Mayotte
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Azadeh Hajmirza
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Éric Bonneil
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - François Béliveau
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada
| | - Sybille Pfammatter
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Albert Feghaly
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Geneviève Boucher
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Patrick Gendron
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Pierre Thibault
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Department of Chemistry, Faculty of Arts and Science, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Frédéric Barabé
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Sébastien Lemieux
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| | - Guillaume Richard-Carpentier
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medicine, Division of Medical Oncology and Hematology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Josée Hébert
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada.
| | - Vincent-Philippe Lavallée
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada; Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Hematology and Oncology Division, Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC H3T 1C5, Canada.
| | - Philippe P Roux
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| | - Guy Sauvageau
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada.
| |
Collapse
|
3
|
Stolpa W, Mizia-Malarz A, Zapała M, Zwiernik B. Can CD34 +CD38 - lymphoblasts, as likely leukemia stem cells, be a prognostic factor in B-cell precursor acute lymphoblastic leukemia in children? Front Pediatr 2023; 11:1213009. [PMID: 37675394 PMCID: PMC10478575 DOI: 10.3389/fped.2023.1213009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/27/2023] [Accepted: 08/01/2023] [Indexed: 09/08/2023] Open
Abstract
Background CD34+CD38- lymphoblasts as likely leukemia stem cells (LSCs) may be responsible for a worse response to treatment and may be a risk factor for recurrence in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Objective The study objective was to assess the prognostic role of CD34+CD38- lymphoblasts in bone marrow on the day of BCP-ALL diagnosis. Methods 115 patients with BCP-ALL, the median age of 4.5 years (range 1.5-17.9 years), gender: female 63 (54.8%) with BCP-ALL were enrolled; Group I (n = 90)-patients with CD34+CD38+ antigens and Group II (n = 20)-patients with CD34+CD38- antigens on the lymphoblast surface. Results A worse response on Days 8, 15, and 33 of therapy and at the end of treatment in Group II (CD34+CD38-) was more often observed but these differences were not statistically significant. A significantly higher incidence of BCP-ALL recurrence was in Group II. Conclusions 1.In BCP-ALL in children, the presence of CD34+CD38- lymphoblasts at the diagnosis does not affect the first remission.2.In BCP-ALL in children, the presence of CD34+CD38- lymphoblasts at the diagnosis may be considered an unfavorable prognostic factor for disease recurrence.3.It is necessary to further search for prognostic factors in BCP-ALL in children.
Collapse
Affiliation(s)
- Weronika Stolpa
- Department of Oncology, Hematology, and Chemotherapy, Upper Silesia Children’s Care Health Centre, Katowice, Poland
| | - Agnieszka Mizia-Malarz
- Department of Oncology, Hematology, and Chemotherapy, Upper Silesia Children’s Care Health Centre, Katowice, Poland
- Department of Pediatrics, Medical University of Silesia, Upper Silesia Children’s Care Health Centre, Katowice, Poland
| | - Magdalena Zapała
- Students’ Research Group, Department of Pediatrics, Medical University of Silesia, Katowice, Poland
| | - Bartosz Zwiernik
- Students’ Research Group, Department of Pediatrics, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
4
|
Bouligny IM, Maher KR, Grant S. Mechanisms of myeloid leukemogenesis: Current perspectives and therapeutic objectives. Blood Rev 2023; 57:100996. [PMID: 35989139 PMCID: PMC10693933 DOI: 10.1016/j.blre.2022.100996] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/23/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic neoplasm which results in clonal proliferation of abnormally differentiated hematopoietic cells. In this review, mechanisms contributing to myeloid leukemogenesis are summarized, highlighting aberrations of epigenetics, transcription factors, signal transduction, cell cycling, and the bone marrow microenvironment. The mechanisms contributing to AML are detailed to spotlight recent findings that convey clinical impact. The applications of current and prospective therapeutic targets are accentuated in addition to reviews of treatment paradigms stratified for each characteristic molecular lesion - with a focus on exploring novel treatment approaches and combinations to improve outcomes in AML.
Collapse
Affiliation(s)
- Ian M Bouligny
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Keri R Maher
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
5
|
Ung J, Tan SF, Fox TE, Shaw JJP, Vass LR, Costa-Pinheiro P, Garrett-Bakelman FE, Keng MK, Sharma A, Claxton DF, Levine RL, Tallman MS, Cabot MC, Kester M, Feith DJ, Loughran TP. Harnessing the power of sphingolipids: Prospects for acute myeloid leukemia. Blood Rev 2022; 55:100950. [PMID: 35487785 PMCID: PMC9475810 DOI: 10.1016/j.blre.2022.100950] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/01/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 11/02/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive, heterogenous malignancy characterized by clonal expansion of bone marrow-derived myeloid progenitor cells. While our current understanding of the molecular and genomic landscape of AML has evolved dramatically and opened avenues for molecularly targeted therapeutics to improve upon standard intensive induction chemotherapy, curative treatments are elusive, particularly in older patients. Responses to current AML treatments are transient and incomplete, necessitating the development of novel treatment strategies to improve outcomes. To this end, harnessing the power of bioactive sphingolipids to treat cancer shows great promise. Sphingolipids are involved in many hallmarks of cancer of paramount importance in AML. Leukemic blast survival is influenced by cellular levels of ceramide, a bona fide pro-death molecule, and its conversion to signaling molecules such as sphingosine-1-phosphate and glycosphingolipids. Preclinical studies demonstrate the efficacy of therapeutics that target dysregulated sphingolipid metabolism as well as their combinatorial synergy with clinically-relevant therapeutics. Thus, increased understanding of sphingolipid dysregulation may be exploited to improve AML patient care and outcomes. This review summarizes the current knowledge of dysregulated sphingolipid metabolism in AML, evaluates how pro-survival sphingolipids promote AML pathogenesis, and discusses the therapeutic potential of targeting these dysregulated sphingolipid pathways.
Collapse
Affiliation(s)
- Johnson Ung
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Su-Fern Tan
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Todd E Fox
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Jeremy J P Shaw
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Experimental Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Luke R Vass
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Experimental Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Pedro Costa-Pinheiro
- Cancer Biology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Francine E Garrett-Bakelman
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Michael K Keng
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Arati Sharma
- Penn State Cancer Institute, Hershey, PA, United States of America
| | - David F Claxton
- Penn State Cancer Institute, Hershey, PA, United States of America
| | - Ross L Levine
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Martin S Tallman
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Mark Kester
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - David J Feith
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Thomas P Loughran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America.
| |
Collapse
|
6
|
Properties of Leukemic Stem Cells in Regulating Drug Resistance in Acute and Chronic Myeloid Leukemias. Biomedicines 2022; 10:biomedicines10081841. [PMID: 36009388 PMCID: PMC9405586 DOI: 10.3390/biomedicines10081841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Notoriously known for their capacity to reconstitute hematological malignancies in vivo, leukemic stem cells (LSCs) represent key drivers of therapeutic resistance and disease relapse, posing as a major medical dilemma. Despite having low abundance in the bulk leukemic population, LSCs have developed unique molecular dependencies and intricate signaling networks to enable self-renewal, quiescence, and drug resistance. To illustrate the multi-dimensional landscape of LSC-mediated leukemogenesis, in this review, we present phenotypical characteristics of LSCs, address the LSC-associated leukemic stromal microenvironment, highlight molecular aberrations that occur in the transcriptome, epigenome, proteome, and metabolome of LSCs, and showcase promising novel therapeutic strategies that potentially target the molecular vulnerabilities of LSCs.
Collapse
|
7
|
Niu J, Peng D, Liu L. Drug Resistance Mechanisms of Acute Myeloid Leukemia Stem Cells. Front Oncol 2022; 12:896426. [PMID: 35865470 PMCID: PMC9294245 DOI: 10.3389/fonc.2022.896426] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/15/2022] [Accepted: 06/06/2022] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukemia (AML) is a polyclonal and heterogeneous hematological malignancy. Relapse and refractory after induction chemotherapy are still challenges for curing AML. Leukemia stem cells (LSCs), accepted to originate from hematopoietic stem/precursor cells, are the main root of leukemogenesis and drug resistance. LSCs are dynamic derivations and possess various elusive resistance mechanisms. In this review, we summarized different primary resistance and remolding mechanisms of LSCs after chemotherapy, as well as the indispensable role of the bone marrow microenvironment on LSCs resistance. Through a detailed and comprehensive review of the spectacle of LSCs resistance, it can provide better strategies for future researches on eradicating LSCs and clinical treatment of AML.
Collapse
Affiliation(s)
| | | | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Synthesis and Anticancer Properties of New 3-Methylidene-1-sulfonyl-2,3-dihydroquinolin-4(1 H)-ones. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27113597. [PMID: 35684532 PMCID: PMC9181899 DOI: 10.3390/molecules27113597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 04/01/2022] [Revised: 04/21/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022]
Abstract
Quinolinones have been known for a long time as broad-spectrum synthetic antibiotics. More recently, the anticancer potential of this group of compounds has been investigated. Following this direction, we obtained a small library of 3-methylidene-1-sulfonyl-2,3-dihydroquinolin-4(1H)-ones with various substituents at positions 1, 2, 6, and 7 of the quinolinone ring system. The cytotoxic activity of the synthesized analogs was tested in the MTT assay on two cancer cell lines in order to determine the structure–activity relationship. All compounds produced high cytotoxic effects in MCF-7, and even higher in HL-60 cells. 2-Ethyl-3-methylidene-1-phenylsulfonyl-2,3-dihydroquinolin-4(1H)-one, which was over 5-fold more cytotoxic for HL-60 than for normal HUVEC cells, was selected for further tests. This analog was shown to inhibit proliferation and induce DNA damage and apoptosis in HL-60 cells.
Collapse
|
9
|
Sui M, Yang H, Guo M, Li W, Gong Z, Jiang J, Li P. Cajanol Sensitizes A2780/Taxol Cells to Paclitaxel by Inhibiting the PI3K/Akt/NF-κB Signaling Pathway. Front Pharmacol 2021; 12:783317. [PMID: 34955854 PMCID: PMC8694871 DOI: 10.3389/fphar.2021.783317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/26/2021] [Accepted: 11/22/2021] [Indexed: 01/06/2023] Open
Abstract
Ovarian cancer is the second most common gynecological malignancy, and one of the most deadly. The bottleneck restricting the treatment of ovarian cancer is its multi-drug resistance to chemotherapy. Cajanol is an isoflavone from pigeon pea (Cajanus cajan) that has been reported to have anti-tumor activity. In this work, we evaluate the effect of cajanol in reversing paclitaxel resistance of the A2780/Taxol ovarian cancer cell line in vitro and in vivo, and we discuss its mechanism of action. We found that 8 μM cajanol significantly restored the sensitivity of A2780/Taxol cells to paclitaxel, and in vivo experiments demonstrated that the combination of 0.5 mM/kg paclitaxel and 2 mM/kg cajanol significantly inhibited the growth of A2780/Taxol metastatic tumors in mice. Flow cytometry, fluorescence quantitative PCR, western blotting and immunohistochemical staining methods were used to study the mechanism of reversing paclitaxel resistance with cajanol. First, we determined that cajanol inhibits paclitaxel efflux in A2780/Taxol cells by down-regulating permeability glycoprotein (P-gp) expression, and further found that cajanol can inhibit P-gp transcription and translation through the PI3K/Akt/NF-κB pathway. The results of this work are expected to provide a new candidate compound for the development of paclitaxel sensitizers.
Collapse
Affiliation(s)
- Ming Sui
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hairong Yang
- Department of Obstetrics and Gynecology, First Hospital of Qiqihar, Qiqihar, China
| | - Mingqi Guo
- Department of Obstetrics and Gynecology, First Hospital of Qiqihar, Qiqihar, China
| | - Wenle Li
- Department of Obstetrics and Gynecology, First Hospital of Qiqihar, Qiqihar, China
| | - Zheng Gong
- Department of Obstetrics and Gynecology, First Hospital of Qiqihar, Qiqihar, China
| | - Jing Jiang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peiling Li
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
10
|
Siedlecka-Kroplewska K, Wrońska A, Kmieć Z. Piceatannol, a Structural Analog of Resveratrol, Is an Apoptosis Inducer and a Multidrug Resistance Modulator in HL-60 Human Acute Myeloid Leukemia Cells. Int J Mol Sci 2021; 22:10597. [PMID: 34638937 PMCID: PMC8509003 DOI: 10.3390/ijms221910597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/05/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Acute myeloid leukemia is characterized by uncontrolled clonal proliferation of abnormal myeloid progenitor cells. Despite recent advances in the treatment of this disease, the prognosis and overall long-term survival for patients remain poor, which drives the search for new chemotherapeutics and treatment strategies. Piceatannol, a polyphenolic compound present in grapes and wine, appears to be a promising chemotherapeutic agent in the treatment of leukemia. The aim of the present study was to examine whether piceatannol induces autophagy and/or apoptosis in HL-60 human acute myeloid leukemia cells and whether HL-60 cells are able to acquire resistance to piceatannol toxicity. We found that piceatannol at the IC90 concentration of 14 µM did not induce autophagy in HL-60 cells. However, it induced caspase-dependent apoptosis characterized by phosphatidylserine externalization, disruption of the mitochondrial membrane potential, caspase-3 activation, internucleosomal DNA fragmentation, PARP1 cleavage, chromatin condensation, and fragmentation of cell nuclei. Our findings also imply that HL-60 cells are able to acquire resistance to piceatannol toxicity via mechanisms related to MRP1 activity. Our results suggest that the use of piceatannol as a potential chemotherapeutic agent may be associated with the risk of multidrug resistance, warranting its use in combination with other chemotherapeutic agents.
Collapse
|
11
|
Yi J, Wang L, Wang XY, Sun J, Yin XY, Hou JX, Chen J, Xie B, Wei HL. Suppression Of Aberrant Activation Of NF-κB Pathway In Drug-resistant Leukemia Stem Cells Contributes To Parthenolide-potentiated Reversal Of Drug Resistance In Leukemia. J Cancer 2021; 12:5519-5529. [PMID: 34405014 PMCID: PMC8364658 DOI: 10.7150/jca.52641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/31/2020] [Accepted: 07/06/2021] [Indexed: 01/05/2023] Open
Abstract
Although many drugs that targeted the specific features of leukemia stem cells (LSCs) have substantial application in the clinical treatment of leukemia, the LSCs relapsed and caused drug-resistant leukemia. Therefore, it is necessary to identify the unique features of LSCs in relapsing and drug-resistant leukemia and also to explore the drugs that directed at these features. Our clinical data have indicated that relapsed patients with acute myeloid leukemia have more abundant proportion of LSCs with enhanced breast cancer resistance protein (BCRP) and P-glycoprotein (P-gp) expression when compared to the untreated patients. The results showed that compared with LSCs derived from sensitive K562 cells, LSCs from drug-resistant K562/ADM cells have much higher chemotherapeutic resistance, and so we termed these cells as “drug-resistant LSCs”. Subsequently, aberrant activation of NF-κB pathway in drug-resistant LSCs was further using gene chip analysis. Also, parthenolide (PTL), which is a specific NF-κB inhibitor, effectively eliminated drug-resistant LSCs and enhanced the sensitivity of K562/ADM cells to doxorubicin-induced apoptosis by down-regulating NF-κB pathway-mediated P-gp expression. These findings make the research area of LSCs more abundant and provide a potential therapeutic strategy for the treatment of refractory and relapsed leukemia.
Collapse
Affiliation(s)
- Juan Yi
- School of Basic Medical Sciences, Lanzhou University; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 99 Dong Gang West Road, Lanzhou, 730000 Gansu, China
| | - Li Wang
- School of Basic Medical Sciences, Lanzhou University; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 99 Dong Gang West Road, Lanzhou, 730000 Gansu, China
| | - Xiao-Yan Wang
- Gansu Provincial Maternity and Childcare Hospital, 143 North Street, Qi Li He district, Lanzhou, 730050, Gansu, China
| | - Jing Sun
- Lanzhou University Second Hospital, 80 Cui Ying Men, Lin Xia Road, Lanzhou, 730000 Gansu, China
| | - Xiao-Yang Yin
- School of Basic Medical Sciences, Lanzhou University; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 99 Dong Gang West Road, Lanzhou, 730000 Gansu, China
| | - Jin-Xia Hou
- School of Basic Medical Sciences, Lanzhou University; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 99 Dong Gang West Road, Lanzhou, 730000 Gansu, China
| | - Jing Chen
- School of Basic Medical Sciences, Lanzhou University; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 99 Dong Gang West Road, Lanzhou, 730000 Gansu, China
| | - Bei Xie
- School of Basic Medical Sciences, Lanzhou University; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 99 Dong Gang West Road, Lanzhou, 730000 Gansu, China
| | - Hu-Lai Wei
- School of Basic Medical Sciences, Lanzhou University; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, 99 Dong Gang West Road, Lanzhou, 730000 Gansu, China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| |
Collapse
|
12
|
Carter JL, Hege K, Yang J, Kalpage HA, Su Y, Edwards H, Hüttemann M, Taub JW, Ge Y. Targeting multiple signaling pathways: the new approach to acute myeloid leukemia therapy. Signal Transduct Target Ther 2020; 5:288. [PMID: 33335095 PMCID: PMC7746731 DOI: 10.1038/s41392-020-00361-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/24/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults and the second most common form of acute leukemia in children. Despite this, very little improvement in survival rates has been achieved over the past few decades. This is partially due to the heterogeneity of AML and the need for more targeted therapeutics than the traditional cytotoxic chemotherapies that have been a mainstay in therapy for the past 50 years. In the past 20 years, research has been diversifying the approach to treating AML by investigating molecular pathways uniquely relevant to AML cell proliferation and survival. Here we review the development of novel therapeutics in targeting apoptosis, receptor tyrosine kinase (RTK) signaling, hedgehog (HH) pathway, mitochondrial function, DNA repair, and c-Myc signaling. There has been an impressive effort into better understanding the diversity of AML cell characteristics and here we highlight important preclinical studies that have supported therapeutic development and continue to promote new ways to target AML cells. In addition, we describe clinical investigations that have led to FDA approval of new targeted AML therapies and ongoing clinical trials of novel therapies targeting AML survival pathways. We also describe the complexity of targeting leukemia stem cells (LSCs) as an approach to addressing relapse and remission in AML and targetable pathways that are unique to LSC survival. This comprehensive review details what we currently understand about the signaling pathways that support AML cell survival and the exceptional ways in which we disrupt them.
Collapse
Affiliation(s)
- Jenna L Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA.,MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Katie Hege
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jay Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hasini A Kalpage
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yongwei Su
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.,National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA. .,Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA. .,Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
13
|
da Silveira Júnior LS, Soares VDL, Jardim da Silva AS, Gil EA, Pereira de Araújo MDG, Merces Gonçalves CA, Paiva ADS, Moura de Oliveira TM, Oliveira GHDM, Kramer Cavacanti E Silva DG, de Araújo Moura Lemos TM, Moretti Rebecchi IM, de Farias Sales VS, Luchessi AD, Cavalcanti Júnior GB. P-glycoprotein and multidrug resistance-associated protein-1 expression in acute myeloid leukemia: Biological and prognosis implications. Int J Lab Hematol 2020; 42:594-603. [PMID: 32452631 DOI: 10.1111/ijlh.13241] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/23/2020] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Despite the advances in the cure rate for acute myeloid leukemia (AML), a considerable number of patients die from the disease due to the occurrence of multidrug resistance (MDR). Overexpression of the transporter proteins, such as P-glycoprotein (Pgp) and multidrug resistance-associated protein (MRP), confers resistance to the treatment of these leukemias. METHODS To analyze the expression of the Pgp and MRP1 in patients with AML and determine their correlation between expression and demographic, clinical, and laboratorial variables, bone marrow and peripheral blood samples from 346 patients with a diagnosis of AML were assessed for the expression of Pgp and MRP1 by flow cytometry. RESULTS The expression of Pgp and MRP1 was found in 111 (32.1%) and 133 (38.4%) patients, respectively, with greater prevalence in older patients and lower in children, while also observing a high incidence in patients with refractory, recurrence, and secondary disease in comparison with the cases of de novo AML. Regarding the laboratory findings, we observed an association between the expression of Pgp and MRP1 and CD34, CD7, and also M7, M5a, and M2-AML of French-American-British classification. CONCLUSIONS The results showed that the detection of MDR phenotype by flow cytometry can be a molecular marker for prognosis of patients with AML.
Collapse
Affiliation(s)
| | - Victor de Lima Soares
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil
| | - Alessandra Suelen Jardim da Silva
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil.,Faculdade de Ciências da Saúde do Trairi/UFRN, Santa Cruz, Brazil
| | - Erica Aires Gil
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil
| | - Maria das Graças Pereira de Araújo
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil.,Laboratório de Citometria de Fluxo, Hemocentro Dalton Cunha, Natal, Brazil
| | - Ciro Alexandre Merces Gonçalves
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil.,Laboratório de Citometria de Fluxo, Hemocentro Dalton Cunha, Natal, Brazil
| | - Aldair de Souza Paiva
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil
| | - Taissa Maria Moura de Oliveira
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil
| | | | | | | | | | - Valeria Soraya de Farias Sales
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil
| | - André Ducati Luchessi
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil
| | - Geraldo Barroso Cavalcanti Júnior
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte (DACT/UFRN), Natal, Brazil.,Laboratório de Citometria de Fluxo, Hemocentro Dalton Cunha, Natal, Brazil
| |
Collapse
|
14
|
New uracil analogs as downregulators of ABC transporters in 5-fluorouracil-resistant human leukemia HL-60 cell line. Mol Biol Rep 2019; 46:5831-5839. [DOI: 10.1007/s11033-019-05017-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2019] [Accepted: 07/31/2019] [Indexed: 12/30/2022]
Abstract
AbstractOverexpression of ATP-binding cassette (ABC) transporters causing multidrug resistance (MDR) in cancer cells is one of the major obstacles in cancer chemotherapy. The 5-FU resistant subclone (HL-60/5FU) of the human HL-60 promyelocytic leukemia cell line was selected by the conventional method of continuous exposure of the cells to the drug up to 0.08 mmol/L concentration. HL-60/5FU cells exhibited six-fold enhanced resistance to 5-FU than HL-60 cells. RT-PCR and ELISA assay showed significant overexpression of MDR-related ABC transporters, ABCB1, ABCG2 but especially ABCC1 in the HL-60/5FU as compared with the parental cell line. Three novel synthetic 5-methylidenedihydrouracil analogs, U-236, U-332 and U-359, selected as highly cytotoxic for HL-60 cells in MTT test, showed similar cytotoxicity in the resistant cell line. When co-incubated with 5-FU, these analogs were found to down-regulate the expression of all three transporters. However, the most pronounced effect was caused by U-332 which almost completely abolished ABCC1 expression in the resistant HL-60/5FU cells. Additionally, U-332 inhibited the activity of ATPase, an enzyme which catalyzes hydrolysis of ATP, providing energy to efflux drugs from the cells through the cellular membranes. Taken together, the obtained data suggest that acquired 5-FU resistance in HL-60/5FU cells results from overexpression of ABCC1 and that targeting ABCC1 expression could be a potential approach to re-sensitize resistant leukemia cells to 5-FU. The synthetic uracil analog U-332, which can potently down-regulate ABC transporter expression and therefore disturb drug efflux, can be considered an efficient ABCC1 regulator in cancer cells.
Collapse
|
15
|
Mastelaro de Rezende M, Ferreira AT, Paredes-Gamero EJ. Leukemia stem cell immunophenotyping tool for diagnostic, prognosis, and therapeutics. J Cell Physiol 2019; 235:4989-4998. [PMID: 31709540 DOI: 10.1002/jcp.29394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/07/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022]
Abstract
The existence of cancer stem cells is debatable in numerous solid tumors, yet in leukemia, there is compelling evidence of this cell population. Leukemic stem cells (LSCs) are altered cells in which accumulating genetic and/or epigenetic alterations occur, resulting in the transition between the normal, preleukemic, and leukemic status. These cells do not follow the normal differentiation program; they are arrested in a primitive state but with high proliferation potential, generating undifferentiated blast accumulation and a lack of a mature cell population. The identification of LSCs might guide stem cell biology research and provide key points of distinction between these cells and their normal counterparts. The identification and characterization of the main features of LSCs can be useful as tools for diagnosis and treatment. In this context, the aim of the present review was to connect immunophenotype data in the main types of leukemia to further guide technical improvements.
Collapse
Affiliation(s)
| | - Alice T Ferreira
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Edgar J Paredes-Gamero
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,Division - Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| |
Collapse
|
16
|
Siedlecka-Kroplewska K, Ślebioda T, Kmieć Z. Induction of autophagy, apoptosis and aquisition of resistance in response to piceatannol toxicity in MOLT-4 human leukemia cells. Toxicol In Vitro 2019; 59:12-25. [DOI: 10.1016/j.tiv.2019.03.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/21/2019] [Revised: 03/09/2019] [Accepted: 03/29/2019] [Indexed: 01/09/2023]
|
17
|
Chk1 Inhibitor MK-8776 Restores the Sensitivity of Chemotherapeutics in P-glycoprotein Overexpressing Cancer Cells. Int J Mol Sci 2019; 20:ijms20174095. [PMID: 31443367 PMCID: PMC6747525 DOI: 10.3390/ijms20174095] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/05/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022] Open
Abstract
P-glycoprotein (P-gp), which is encoded by the ATP-binding cassette (ABC) transporter subfamily B member 1 (ABCB1) gene, is one of the most pivotal ABC transporters that transport its substrates across the cell membrane. Its overexpression is one of the confirmed causes of multidrug resistance (MDR), which results in the failure of cancer treatment. Here, we report that checkpoint kinase (Chk) 1 inhibitor MK-8776, a drug candidate in clinical trial, can restore the sensitivity of chemotherapeutics that are substrates of P-gp in KB-C2, SW620/Ad300 cells and human embryonic kidney (HEK)293/ABCB1 cells that overexpress P-gp. MK-8776 remarkably enhanced the cellular [3H]-paclitaxel accumulation and suppressed the efflux function of P-gp without reducing its expression and affecting its cellular localization in cancer cells. Furthermore, MK-8776 (0–40 μM) stimulated the activity of ATPase in P-gp, which was 4.1-fold greater than the control. In addition, MK-8776 formed a cation–π bond and π–π interaction with key residues of the substrate-binding site in P-gp, as indicated by computer-aided molecular docking study. Our study indicated that MK-8776 may significantly enhance the sensitivity of chemotherapeutics that are substrates of P-gp, providing important information for its application in the reversal of MDR.
Collapse
|
18
|
Panyajai P, Tima S, Chiampanichayakul S, Anuchapreeda S. Dietary Turmeric Bisdemethoxycurcumin Suppresses Wilms’ Tumor 1 and CD34 Protein Expressions in KG-1a Leukemic Stem Cells. Nutr Cancer 2019; 71:1189-1200. [DOI: 10.1080/01635581.2019.1598565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/03/2023]
Affiliation(s)
- Pawaret Panyajai
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Singkome Tima
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand
| | - Sawitree Chiampanichayakul
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Songyot Anuchapreeda
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
19
|
Guo L, Chen L, Wang H. CD45 correlates with adverse risk stratification, decreased treatment response and unfavorable survival profiles in elderly acute myeloid leukemia patients. Cancer Biomark 2018; 23:455-463. [DOI: 10.3233/cbm-181602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/12/2023]
|
20
|
Cytokeratin 19 (KRT19) has a Role in the Reprogramming of Cancer Stem Cell-Like Cells to Less Aggressive and More Drug-Sensitive Cells. Int J Mol Sci 2018; 19:ijms19051423. [PMID: 29747452 PMCID: PMC5983664 DOI: 10.3390/ijms19051423] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/15/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023] Open
Abstract
Cytokeratin 19 (KRT19) is a cytoplasmic intermediate filament protein, which is responsible for structural rigidity and multipurpose scaffolds. In several cancers, KRT19 is overexpressed and may play a crucial role in tumorigenic transformation. In our previous study, we revealed the role of KRT19 as signaling component which mediated Wnt/NOTCH crosstalk through NUMB transcription in breast cancer. Here, we investigated the function of KRT19 in cancer reprogramming and drug resistance in breast cancer cells. We found that expression of KRT19 was attenuated in several patients-derived breast cancer tissues and patients with a low expression of KRT19 were significantly correlated with poor prognosis in breast cancer patients. Consistently, highly aggressive and drug-resistant breast cancer patient-derived cancer stem cell-like cells (konkuk university-cancer stem cell-like cell (KU-CSLCs)) displayed higher expression of cancer stem cell (CSC) markers, including ALDH1, CXCR4, and CD133, but a much lower expression of KRT19 than that is seen in highly aggressive triple negative breast cancer MDA-MB231 cells. Moreover, we revealed that the knockdown of KRT19 in MDA-MB231 cells led to an enhancement of cancer properties, such as cell proliferation, sphere formation, migration, and drug resistance, while the overexpression of KRT19 in KU-CSLCs resulted in the significant attenuation of cancer properties. KRT19 regulated cancer stem cell reprogramming by modulating the expression of cancer stem cell markers (ALDH1, CXCR4, and CD133), as well as the phosphorylation of Src and GSK3β (Tyr216). Therefore, our data may imply that the modulation of KRT19 expression could be involved in cancer stem cell reprogramming and drug sensitivity, which might have clinical implications for cancer or cancer stem cell treatment.
Collapse
|
21
|
Arrigoni E, Del Re M, Galimberti S, Restante G, Rofi E, Crucitta S, Baratè C, Petrini M, Danesi R, Di Paolo A. Concise Review: Chronic Myeloid Leukemia: Stem Cell Niche and Response to Pharmacologic Treatment. Stem Cells Transl Med 2018; 7:305-314. [PMID: 29418079 PMCID: PMC5827745 DOI: 10.1002/sctm.17-0175] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2017] [Accepted: 01/09/2018] [Indexed: 12/27/2022] Open
Abstract
Nowadays, more than 90% of patients affected by chronic myeloid leukemia (CML) survive with a good quality of life, thanks to the clinical efficacy of tyrosine kinase inhibitors (TKIs). Nevertheless, point mutations of the ABL1 pocket occurring during treatment may reduce binding of TKIs, being responsible of about 20% of cases of resistance among CML patients. In addition, the presence of leukemic stem cells (LSCs) represents the most important event in leukemia progression related to TKI resistance. LSCs express stem cell markers, including active efflux pumps and genetic and epigenetic alterations together with deregulated cell signaling pathways involved in self-renewal, such as Wnt/β-catenin, Notch, and Hedgehog. Moreover, the interaction with the bone marrow microenvironment, also known as hematopoietic niche, may influence the phenotype of surrounding cells, which evade mechanisms controlling cell proliferation and are less sensitive or frankly resistant to TKIs. This Review focuses on the role of LSCs and stem cell niche in relation to response to pharmacological treatments. A literature search from PubMed database was performed until April 30, 2017, and it has been analyzed according to keywords such as chronic myeloid leukemia, stem cell, leukemic stem cells, hematopoietic niche, tyrosine kinase inhibitors, and drug resistance. Stem Cells Translational Medicine 2018;7:305-314.
Collapse
Affiliation(s)
- Elena Arrigoni
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Sara Galimberti
- Unit of Hematology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giuliana Restante
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Eleonora Rofi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Claudia Baratè
- Unit of Hematology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Mario Petrini
- Unit of Hematology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Antonello Di Paolo
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
22
|
Salerno L, Romeo G, Modica MN, Amata E, Sorrenti V, Barbagallo I, Pittalà V. Heme oxygenase-1: A new druggable target in the management of chronic and acute myeloid leukemia. Eur J Med Chem 2017; 142:163-178. [DOI: 10.1016/j.ejmech.2017.07.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/01/2017] [Revised: 07/14/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
|
23
|
Chen J, Wei H, Cheng J, Xie B, Wang B, Yi J, Tian B, Liu Z, Wang F, Zhang Z. Characteristics of doxorubicin-selected multidrug-resistant human leukemia HL-60 cells with tolerance to arsenic trioxide and contribution of leukemia stem cells. Oncol Lett 2017; 15:1255-1262. [PMID: 29399180 DOI: 10.3892/ol.2017.7353] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/29/2015] [Accepted: 06/09/2017] [Indexed: 01/26/2023] Open
Abstract
The present study selected and characterized a multidrug-resistant HL-60 human acute promyelocytic leukemia cell line, HL-60/RS, by exposure to stepwise incremental doses of doxorubicin. The drug-resistant HL-60/RS cells exhibited 85.68-fold resistance to doxorubicin and were cross-resistant to other chemotherapeutics, including cisplatin, daunorubicin, cytarabine, vincristine and etoposide. The cells over-expressed the transporters P-glycoprotein, multidrug-resistance-related protein 1 and breast-cancer-resistance protein, encoded by the adenosine triphosphate-binding cassette (ABC)B1, ABCC1 and ABCG2 genes, respectively. Unlike other recognized chemoresistant leukemia cell lines, HL-60/RS cells were also strongly cross-resistant to arsenic trioxide. The proportion of leukemia stem cells (LSCs) increased synchronously with increased of drug resistance in the doxorubicin-induced HL-60 cell population. The present study confirmed that doxorubicin-induced HL-60 cells exhibited multidrug-resistance and high arsenic-trioxide resistance. Drug-resistance in these cells may be due to surviving chemoresistant LSCs in the HL-60 population, which have been subjected to long and consecutive selection by doxorubicin.
Collapse
Affiliation(s)
- Jing Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hulai Wei
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jie Cheng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Bei Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Bei Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Juan Yi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Baoying Tian
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhuan Liu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Feifei Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhewen Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
24
|
Paprocka M, Bielawska-Pohl A, Rossowska J, Krawczenko A, Duś D, Kiełbiński M, Haus O, Podolak-Dawidziak M, Kuliczkowski K. MRP1 protein expression in leukemic stem cells as a negative prognostic marker in acute myeloid leukemia patients. Eur J Haematol 2017; 99:415-422. [DOI: 10.1111/ejh.12938] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 08/02/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Maria Paprocka
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Aleksandra Bielawska-Pohl
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Agnieszka Krawczenko
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Danuta Duś
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Marek Kiełbiński
- Department and Clinic of Hematology; Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| | - Olga Haus
- Department and Clinic of Hematology; Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| | - Maria Podolak-Dawidziak
- Department and Clinic of Hematology; Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| | - Kazimierz Kuliczkowski
- Department and Clinic of Hematology; Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| |
Collapse
|
25
|
The Effects of Rutin on the Gene Expression of Dazl, Bcl2, and Caspase3 in Idarubicin-induced Testicular Damages in Mice. IRANIAN RED CRESCENT MEDICAL JOURNAL 2017. [DOI: 10.5812/ircmj.44765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
|
26
|
Morad SAF, Davis TS, MacDougall MR, Tan SF, Feith DJ, Desai DH, Amin SG, Kester M, Loughran TP, Cabot MC. Role of P-glycoprotein inhibitors in ceramide-based therapeutics for treatment of cancer. Biochem Pharmacol 2017; 130:21-33. [PMID: 28189725 DOI: 10.1016/j.bcp.2017.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/04/2016] [Accepted: 02/01/2017] [Indexed: 10/20/2022]
Abstract
The anticancer properties of ceramide, a sphingolipid with potent tumor-suppressor properties, can be dampened via glycosylation, notably in multidrug resistance wherein ceramide glycosylation is characteristically elevated. Earlier works using the ceramide analog, C6-ceramide, demonstrated that the antiestrogen tamoxifen, a first generation P-glycoprotein (P-gp) inhibitor, blocked C6-ceramide glycosylation and magnified apoptotic responses. The present investigation was undertaken with the goal of discovering non-anti-estrogenic alternatives to tamoxifen that could be employed as adjuvants for improving the efficacy of ceramide-centric therapeutics in treatment of cancer. Herein we demonstrate that the tamoxifen metabolites, desmethyltamoxifen and didesmethyltamoxifen, and specific, high-affinity P-gp inhibitors, tariquidar and zosuquidar, synergistically enhanced C6-ceramide cytotoxicity in multidrug resistant HL-60/VCR acute myelogenous leukemia (AML) cells, whereas the selective estrogen receptor antagonist, fulvestrant, was ineffective. Active C6-ceramide-adjuvant combinations elicited mitochondrial ROS production and cytochrome c release, and induced apoptosis. Cytotoxicity was mitigated by introduction of antioxidant. Effective adjuvants markedly inhibited C6-ceramide glycosylation as well as conversion to sphingomyelin. Active regimens were also effective in KG-1a cells, a leukemia stem cell-like line, and in LoVo human colorectal cancer cells, a solid tumor model. In summary, our work details discovery of the link between P-gp inhibitors and the regulation and potentiation of ceramide metabolism in a pro-apoptotic direction in cancer cells. Given the active properties of these adjuvants in synergizing with C6-ceramide, independent of drug resistance status, stemness, or cancer type, our results suggest that the C6-ceramide-containing regimens could provide alternative, promising therapeutic direction, in addition to finding novel, off-label applications for P-gp inhibitors.
Collapse
Affiliation(s)
- Samy A F Morad
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Greenville, NC, United States; Department of Pharmacology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Traci S Davis
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Greenville, NC, United States
| | - Matthew R MacDougall
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Greenville, NC, United States
| | - Su-Fern Tan
- Department of Medicine, Hematology/Oncology, University of Virginia, Charlottesville, VA, United States
| | - David J Feith
- Department of Medicine, Hematology/Oncology, University of Virginia, Charlottesville, VA, United States; University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, United States
| | - Dhimant H Desai
- Penn State University College of Medicine, Department of Pharmacology, University Drive, Hershey, PA, United States
| | - Shantu G Amin
- Penn State University College of Medicine, Department of Pharmacology, University Drive, Hershey, PA, United States
| | - Mark Kester
- University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, United States
| | - Thomas P Loughran
- Department of Medicine, Hematology/Oncology, University of Virginia, Charlottesville, VA, United States; University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, United States
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Greenville, NC, United States.
| |
Collapse
|
27
|
Song K, Li M, Xu X, Xuan LI, Huang G, Liu Q. Resistance to chemotherapy is associated with altered glucose metabolism in acute myeloid leukemia. Oncol Lett 2016; 12:334-342. [PMID: 27347147 DOI: 10.3892/ol.2016.4600] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/24/2014] [Accepted: 03/08/2016] [Indexed: 12/18/2022] Open
Abstract
Altered glucose metabolism has been described as a cause of chemoresistance in multiple tumor types. The present study aimed to identify the expression profile of glucose metabolism in drug-resistant acute myeloid leukemia (AML) cells and provide potential strategies for the treatment of drug-resistant AML. Bone marrow and serum samples were obtained from patients with AML that were newly diagnosed or had relapsed. The messenger RNA expression of hypoxia inducible factor (HIF)-1α, glucose transporter (GLUT)1, and hexokinase-II was measured by quantitative polymerase chain reaction. The levels of LDH and β subunit of human F1-F0 adenosine triphosphate synthase (β-F1-ATPase) were detected by enzyme-linked immunosorbent and western blot assays. The HL-60 and HL-60/ADR cell lines were used to evaluate glycolytic activity and effect of glycolysis inhibition on cellular proliferation and apoptosis. Drug-resistant HL-60/ADR cells exhibited a significantly increased level of glycolysis compared with the drug-sensitive HL-60 cell line. The expression of HIF-1α, hexokinase-II, GLUT1 and LDH were increased in AML patients with no remission (NR), compared to healthy control individuals and patients with complete remission (CR) and partial remission. The expression of β-F1-ATPase in patients with NR was decreased compared with the expression in the CR group. Treatment of HL-60/ADR cells with 2-deoxy-D-glucose or 3-bromopyruvate increased in vitro sensitivity to Adriamycin (ADR), while treatment of HL-60 cells did not affect drug cytotoxicity. Subsequent to treatment for 24 h, apoptosis in these two cell lines showed no significant difference. However, glycolytic inhibitors in combination with ADR increased cellular necrosis. These findings indicate that increased glycolysis and low efficiency of oxidative phosphorylation may contribute to drug resistance. Targeting glycolysis is a viable strategy for modulating chemoresistance in AML.
Collapse
Affiliation(s)
- Kui Song
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, P.R. China; Department of Hematology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528400, P.R. China
| | - Min Li
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Xiaojun Xu
- Department of Hematology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528400, P.R. China; Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - L I Xuan
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Guinian Huang
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
28
|
Khamenehfar A, Gandhi MK, Chen Y, Hogge DE, Li PCH. Dielectrophoretic Microfluidic Chip Enables Single-Cell Measurements for Multidrug Resistance in Heterogeneous Acute Myeloid Leukemia Patient Samples. Anal Chem 2016; 88:5680-8. [PMID: 27149245 DOI: 10.1021/acs.analchem.5b04446] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
The front-line treatment for adult acute myeloid leukemia (AML) is anthracycline-based combination chemotherapy. However, treatment outcomes remain suboptimal with relapses frequently observed. Among the mechanisms of treatment failure is multidrug resistance (MDR) mediated by the ABCB1, ABCC1, and ABCG2 drug-efflux transporters. Although genetic and phenotypic heterogeneity between leukemic blast cells is a well-recognized phenomenon, there remains minimal data on differences in MDR activity at the individual cell level. Specifically, functional assays that can distinguish the variability in MDR activity between individual leukemic blasts are lacking. Here, we outline a new dielectrophoretic (DEP) chip-based assay. This assay permits measurement of drug accumulation in single cells, termed same-single-cell analysis in the accumulation mode (SASCA-A). Initially, the assay was optimized in pretherapy samples from 20 adults with AML whose leukemic blasts had MDR activity against the anthracyline daunorubicin (DNR) tested using multiple MDR inhibitors. Parameters tested were initial drug accumulation, time to achieve signal saturation, fold-increase of DNR accumulation with MDR inhibition, ease of cell trapping, and ease of maintaining the trapped cells stationary. This enabled categorization into leukemic blast cells with MDR activity (MDR(+)) and leukemic blast cells without MDR activity (MDR(-ve)). Leukemic blasts could also be distinguished from benign white blood cells (notably these also lacked MDR activity). MDR(-ve) blasts were observed to be enriched in samples taken from patients who went on to enter complete remission (CR), whereas MDR(+) blasts were frequently observed in patients who failed to achieve CR following front-line chemotherapy. However, pronounced variability in functional MDR activity between leukemic blasts was observed, with MDR(+) cells not infrequently seen in some patients that went on to achieve CR. Next, we tested MDR activity in two paired AML patient samples. Pretherapy samples taken from patients that achieved CR to front-line chemotherapy were compared with samples taken at time of subsequent relapse. MDR(+) cells were frequently observed in leukemic blast cells in both pretherapy and relapsed samples, consistent with MDR as a mechanism of relapse in these patients. We demonstrate the ability of a new DEP microfluidic chip-based assay to identify heterogeneity in MDR activity in leukemic blasts. The test provides a platform for future studies to characterize the mechanistic basis for heterogeneity in MDR activity at the individual cell level.
Collapse
Affiliation(s)
| | - Maher K Gandhi
- The University of Queensland , Diamantina Institute, 37 Kent Street, Woolloongabba, Queensland, Australia
| | | | - Donna E Hogge
- Terry Fox Laboratory, BC Cancer Agency , 675 West 10th Avenue, Vancouver, British Columbia, Canada
| | | |
Collapse
|
29
|
Zhao W, Liu S, Dou Q, Li C, DU J, Ren W. The role and mechanism of WEE1 on the cisplatin resistance reversal of the HepG2/DDP human hepatic cancer cell line. Oncol Lett 2015; 10:3081-3086. [PMID: 26722293 DOI: 10.3892/ol.2015.3647] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/20/2014] [Accepted: 05/20/2015] [Indexed: 12/12/2022] Open
Abstract
Drug resistance to cisplatin with continuous drug treatment is one of the most common causes of chemotherapy failure in hepatic carcinoma. Accumulating evidence suggests that WEE1 G2 checkpoint kinase (WEE1) is involved in cisplatin resistance, which has been demonstrated to correlate with cancer initiation and progression. However, the role and molecular mechanism of WEE1 in the drug resistance of hepatic cancer remains unclear. In the present study, using the WEE-knockdown hepatic cancer cell line HepG2/DDP, the role of WEE1 and its molecular mechanism were investigated. It was demonstrated that silencing WEE1 expression resulted in an increased cisplatin sensitivity of HepG2/DDP, in addition to an increased rate of apoptosis and intracellular concentration of rhodamine 123. The expression levels of P-gp, MDR1, MRP1, LRP, BCL-2, survivin and GST in WEE1-silenced HepG2/DDP cells were significantly reduced, and phosphorylation levels of MEK and ERK were significantly downregulated. The results demonstrated that WEE1 negatively regulated the multidrug resistance potential of human hepatic cancer cells by modulating the expression of relevant drug resistance genes and the activity of the MEK/ERK pathway. Therefore, WEE1 may be a monitoring bio-marker for drug resistance, and a therapeutic target in hepatic cancer.
Collapse
Affiliation(s)
- Weifeng Zhao
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Shuyuan Liu
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Qian Dou
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Changan Li
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jingpei DU
- Department of Infections, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Weihua Ren
- Central Laboratory, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| |
Collapse
|
30
|
Andrews TE, Wang D, Harki DA. Cell surface markers of cancer stem cells: diagnostic macromolecules and targets for drug delivery. Drug Deliv Transl Res 2015; 3:121-42. [PMID: 25787981 DOI: 10.1007/s13346-012-0075-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/27/2022]
Abstract
The recognition that the persistence of cancer stem cells (CSCs) in patients following chemotherapy can result in disease relapse underscores the necessity to develop therapeutics against those cells. CSCs display a unique repertoire of cell surface macromolecules, which have proven essential for their characterization and isolation. Additionally, CSC-specific cell surface macromolecules or markers provide targets for the development of specific agents to destroy them. In this review, we compiled those cell surface molecules that have been validated as CSC markers for many common blood and solid tumors. We describe the unique chemical and structural features of the most common cell surface markers, as well as recent efforts to deliver chemotherapeutic agents into CSCs by targeting those macromolecules.
Collapse
Affiliation(s)
- Timothy E Andrews
- Department of Medicinal Chemistry, University of Minnesota, 717 Delaware St SE, Minneapolis, MN, 55414, USA
| | | | | |
Collapse
|
31
|
Voso MT, Santini V, Fabiani E, Fianchi L, Criscuolo M, Falconi G, Guidi F, Hohaus S, Leone G. Why methylation is not a marker predictive of response to hypomethylating agents. Haematologica 2015; 99:613-9. [PMID: 24688109 DOI: 10.3324/haematol.2013.099549] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022] Open
Abstract
The azanucleotides azacitidine and decitabine have been shown to induce hematologic response and prolong survival in higher-risk myelodysplastic syndromes. They are inhibitors of DNA methyltransferase-1 and induce DNA-hypomethylation. Induction of apoptosis is also clinically relevant, in particular during the first treatment cycles, when cytopenia is a frequent side-effect. Since the hypomethylating effect is reversible, and the malignant clone has been shown to persist in most responding patients, several cycles are necessary to achieve and maintain responses, while treatment interruption is associated with rapid relapse. Methylation studies have shown global and gene-specific hypermethylation in myelodysplastic syndromes, but there seems to be little relation between the degree of demethylation following hypomethylating treatment and hematologic response. The presence of concurrent genomic hypermethylation and hypomethylation may impair the predictive power of current detection techniques. This scenario has been complicated by the identification of epigenetic enzyme mutations, including TET2, IDH1/2, DNMT3A and EZH2, which are important for response to hypomethylating treatment. Changes in azanucleotide metabolism genes may also play a role. In the future, methylation analysis concentrating not only on promoters, but also on gene bodies and intergenic regions, may identify key genes in patients with the highest probability of response to azanucleotides and allow a patient-tailored approach.
Collapse
|
32
|
Zhao W, Chen R, Zhao M, Li L, Fan L, Che XM. High glucose promotes gastric cancer chemoresistance in vivo and in vitro. Mol Med Rep 2015; 12:843-50. [PMID: 25815791 PMCID: PMC4438965 DOI: 10.3892/mmr.2015.3522] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/03/2014] [Accepted: 12/02/2014] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to determine whether gastric cancer chemoresistance was increased under high glucose conditions by means of a clinical case study and experimental cytology. The expression of nicotinamide phosphoribosyltransferase (Nampt), silent information regulator 1 (sirt1), p53, p-glycoprotein (P-gp) and topoisomerase (topo)-IIα was evaluated in gastric cancer tissues and gastric cancer with diabetes tissues by immunohistochemistry. Subsequently, the survival time of the patients was assessed. For further investigation, the human gastric cancer cell line SGC7901 was subjected to different glucose concentrations and the aforementioned proteins were detected using reverse transcription-quantitative polymerase chain reaction and western blot analysis. Finally, cell sensitivity to chemotherapy treatment was examined in order to elucidate the role of high glucose in MDR. Positive expression of Nampt, Sirt1, p53, P-gp and Topo-IIα was observed to be higher in gastric cancer with diabetes patients compared with gastric cancer patients (P=0.01, 0.003, 0.0025, 0.016 and 0.336, respectively) with reduced survival time. Similar results were observed in SGC7901 cells. Additionally, cell proliferation rates of SGC7901 cells increased at glucose concentrations of 4,500 and 9,000 mg/l. Notably, the inhibition rates of 5-fluorouracil on cells decreased over 48 h when treated with 4,500 and 9,000 mg/l glucose compared with 1,000 mg/l. In conclusion, patients suffering from gastric cancer and diabetes exhibited greater negative effects, such as a poorer response to chemotherapy and had a lower survival time. High glucose conditions promoted gastric cancer cell proliferation and reduced susceptibility to chemotherapy drugs. These data provided a potential diagnostic and therapeutic strategy for gastric cancer chemoresistance.
Collapse
Affiliation(s)
- Wei Zhao
- Department of General Surgery, The First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Rui Chen
- Department of Neonatal Surgery, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Mei Zhao
- Department of Pharmacology, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Liang Li
- Department of General Surgery, The First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lin Fan
- Department of General Surgery, The First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiang-Ming Che
- Department of General Surgery, The First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
33
|
Zhang Y, Chen HX, Zhou SY, Wang SX, Zheng K, Xu DD, Liu YT, Wang XY, Wang X, Yan HZ, Zhang L, Liu QY, Chen WQ, Wang YF. Sp1 and c-Myc modulate drug resistance of leukemia stem cells by regulating survivin expression through the ERK-MSK MAPK signaling pathway. Mol Cancer 2015; 14:56. [PMID: 25890196 PMCID: PMC4357193 DOI: 10.1186/s12943-015-0326-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/06/2014] [Accepted: 02/23/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is initiated and maintained by a subset of self-renewing leukemia stem cells (LSCs), which contribute to the progression, recurrence and therapeutic resistance of leukemia. However, the mechanisms underlying the maintenance of LSCs drug resistance have not been fully defined. In this study, we attempted to elucidate the mechanisms of LSCs drug resistance. METHODS We performed reverse phase protein arrays to analyze the expression of anti-apoptotic proteins in the LSC-enriched leukemia cell line KG-1a. Immuno-blotting, cell viability and clinical AML samples were evaluated to verify the micro-assay results. The characteristics and transcriptional regulation of survivin were analyzed with the relative luciferase reporter assay, mutant constructs, chromatin immuno-precipitation (ChIP), quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), and western blotting. The levels of Sp1, c-Myc, phospho-extracellular signal-regulated kinase (p-ERK), phospho-mitogen and stress-activated protein kinase (p-MSK) were investigated in paired CD34+ and CD34- AML patient samples. RESULTS Survivin was highly over-expressed in CD34 + CD38- KG-1a cells and paired CD34+ AML patients compared with their differentiated counterparts. Functionally, survivin contributes to the drug resistance of LSCs, and Sp1 and c-Myc concurrently regulate levels of survivin transcription. Clinically, Sp1 and c-Myc were significantly up-regulated and positively correlated with survivin in CD34+ AML patients. Moreover, Sp1 and c-Myc were further activated by the ERK/MSK mitogen-activated protein kinase (MAPK) signaling pathway, modulating survivin levels. CONCLUSION Our findings demonstrated that ERK/MSK/Sp1/c-Myc axis functioned as a critical regulator of survivin expression in LSCs, offering a potential new therapeutic strategy for LSCs therapy.
Collapse
Affiliation(s)
- Yi Zhang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Hai-xuan Chen
- College of Medicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Shu-yan Zhou
- Department of Pathological Physiology, Wan-nan Medical College, 241000, Wuhu, P.R China.
| | - Shao-xiang Wang
- College of Medicine, Shenzhen University, 518020, Shenzhen, P.R China.
| | - Kai Zheng
- College of Medicine, Shenzhen University, 518020, Shenzhen, P.R China.
| | - Dan-dan Xu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Yu-ting Liu
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Xiao-yan Wang
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Xiao Wang
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Hai-Zhao Yan
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Li Zhang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
| | - Qiu-ying Liu
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Wan-qun Chen
- College of Medicine, Jinan University, 510632, Guangzhou, P.R China.
| | - Yi-fei Wang
- College of Life Science and Technology, Jinan University, 510632, Guangzhou, P.R, China.
- Institute of Biomedicine, Jinan University, 510632, Guangzhou, P.R China.
| |
Collapse
|
34
|
Progress in RNAi-mediated Molecular Therapy of Acute and Chronic Myeloid Leukemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e240. [DOI: 10.1038/mtna.2015.13] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/13/2015] [Accepted: 03/26/2015] [Indexed: 02/08/2023]
|
35
|
Basseville A, Robey RW, Bahr JC, Bates SE. Breast Cancer Resistance Protein (BCRP) or ABCG2. DRUG TRANSPORTERS 2014:187-221. [DOI: 10.1002/9781118705308.ch11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/03/2025]
|
36
|
Zhang H, Fang H, Wang K. Reactive oxygen species in eradicating acute myeloid leukemic stem cells. Stem Cell Investig 2014; 1:13. [PMID: 27358859 DOI: 10.3978/j.issn.2306-9759.2014.04.03] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/07/2014] [Accepted: 04/20/2014] [Indexed: 12/13/2022]
Abstract
Leukemic stem cells (LSCs) have been proven to drive leukemia initiation, progression and relapse, and are increasingly being used as a critical target for therapeutic intervention. As an essential feature in LSCs, reactive oxygen species (ROS) homeostasis has been extensively exploited in the past decade for targeting LSCs in acute myeloid leukemia (AML). Most, if not all, agents that show therapeutic benefits are able to alter redox status by inducing ROS, which confers selectivity in eradicating AML stem cells but sparing normal counterparts. In this review, we provide the comprehensive update of ROS-generating agents in the context of their impacts on our understanding of the pathogenesis of AML and its therapy. We anticipate that further characterizing these ROS agents will help us combat against AML in the coming era of LSC-targeting strategy.
Collapse
Affiliation(s)
- Hui Zhang
- 1 State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China ; 2 Pediatric department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Hai Fang
- 1 State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China ; 2 Pediatric department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Kankan Wang
- 1 State Key Laboratory of Medical Genomics and Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China ; 2 Pediatric department, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
37
|
Ahlers J, Witte KE, Schwarze CP, Lang P, Handgretinger R, Ebinger M. Therapy response correlates with ALDH activity in ALDH low-positive childhood acute lymphoblastic leukemias. Pediatr Hematol Oncol 2014; 31:303-10. [PMID: 24308780 DOI: 10.3109/08880018.2013.859189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022]
Abstract
The malignant cells of childhood acute lymphoblastic leukemia (ALL) do not form a homogenous entity but a collection of differently maturated blasts. The most immature leukemia cells may be more resistant to therapy than the bulk of more differentiated blasts. We studied 42 patients with childhood ALL treated according to the ALL-BFM 2000 protocol. At diagnosis, we determined the immunophenotype and the aldehyde dehydrogenase (ALDH) activity of the leukemic cells. Additionally, we investigated the expression of CD34, CD38 and CD45 to define a population of immunophenotypically immature cells (CD34(+)/CD38(-)/CD45(-/low)). We then studied levels of minimal residual disease (MRD) after induction therapy (day 33) to determine therapy response. Including all cases (n = 42), there was no correlation between ALDH positive cells, CD34(+)/CD38(-)/CD45(-/low) cells and MRD levels. A subset of 18 ALLs displayed a more mature phenotype with low-ALDH positivity (< 1%). Analyzing this cohort, ALDH positive blasts overlapped with the CD34(+)/CD38(-)/CD45(-/low) population. The initial rate of ALDH positivity correlated with MRD levels at day 33 of therapy (r = 0.61, P < .01). We conclude that in pediatric ALL, ALDH positivity as a marker of immaturity and stemness has prognostic significance only in phenotypically mature cases when the ALDH activity is not a property of the majority of the leukemic blasts. In case of an immature ALL phenotype, ALDH activity might be an inherent characteristic of the whole leukemia and is not limited to a more immature subpopulation that could confer to resistance and increased MRD-levels during therapy.
Collapse
Affiliation(s)
- Jörg Ahlers
- Klinikum Chemnitz, Children's Hospital , Chemnitz , Germany
| | | | | | | | | | | |
Collapse
|
38
|
Low-dose triptolide in combination with idarubicin induces apoptosis in AML leukemic stem-like KG1a cell line by modulation of the intrinsic and extrinsic factors. Cell Death Dis 2013; 4:e948. [PMID: 24309935 PMCID: PMC3877540 DOI: 10.1038/cddis.2013.467] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2013] [Revised: 10/17/2013] [Accepted: 10/21/2013] [Indexed: 01/20/2023]
Abstract
Leukemia stem cells (LSCs) are considered to be the main reason for relapse and are also regarded as a major hurdle for the success of acute myeloid leukemia chemotherapy. Thus, new drugs targeting LSCs are urgently needed. Triptolide (TPL) is cytotoxic to LSCs. Low dose of TPL enhances the cytotoxicity of idarubicin (IDA) in LSCs. In this study, the ability of TPL to induce apoptosis in leukemic stem cell (LSC)-like cells derived from acute myeloid leukemia cell line KG1a was investigated. LSC-like cells sorted from KG1a were subjected to cell cycle analysis and different treatments, and then followed by in vitro methyl thiazole tetrazolium bromide cytotoxicity assay. The effects of different drug combinations on cell viability, intracellular reactive-oxygen species (ROS) activity, colony-forming ability and apoptotic status were also examined. Combination index-isobologram analysis indicates a synergistic effect between TPL and IDA, which inhibits the colony-forming ability of LSC-like cells and induces their apoptosis. We further investigated the expression of Nrf2, HIF-1α and their downstream target genes. LSC-like cells treated with both TPL and IDA have increased levels of ROS, decreased expression of Nrf2 and HIF-1α pathways. Our findings indicate that the synergistic cytotoxicity of TPL and IDA in LSCs-like cells may attribute to both induction of ROS and inhibition of the Nrf2 and HIF-1α pathways.
Collapse
|
39
|
Abstract
The ATP-binding cassette, subfamily G, isoform 2 protein (ABCG2) is an important member of the ABC transporter superfamily, which has been suggested to be involved in multidrug resistance (MDR) in cancer. Its diverse range of substrates includes many common chemotherapeutics such as imatinib, doxorubicin, and mitoxantrone. Physiologically, ABCG2 is highly expressed in areas such as the blood-brain barrier and gastrointestinal tract, where it is thought to play a role in protection against xenobiotic exposure. High ABCG2 expression has also been found in a variety of solid tumors and in hematologic malignancies and has been correlated with poorer clinical outcomes. Furthermore, ABCG2 expression is a characteristic feature of cancer stem cells, which are able to self-renew and differentiate. These cancer stem cells have been postulated to play an important role in MDR, where their inherent ABCG2 expression may allow them to survive chemotherapy and repopulate the tumor after exposure to chemotherapeutics. This observation raises the exciting possibility that by inhibiting ABCG2, cancer stem cells and other cancers may be targeted and eradicated, at which point conventional chemotherapeutics would be sufficient to eliminate the remaining tumor cells. Inhibitors of ABCG2, such as tyrosine kinase inhibitors, phosphodiesterase-5 inhibitors, and the fumitremorgin-type indolyl diketopiperazine, Ko143 [(3S,6S,12aS)-1,2,3,4,6,7,12,12a-octahydro-9-methoxy-6-(2-methylpropyl)-1,4-dioxopyrazino[1',2':1,6]pyrido[3,4-b]indole-3-propanoic acid 1,1-dimethylethyl ester], could potentially be used for this purpose. However, these agents are still awaiting comprehensive clinical assessment.
Collapse
Affiliation(s)
- Alexandra E Stacy
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | | | | |
Collapse
|
40
|
FLT3-ITD and MLL-PTD influence the expression of MDR-1, MRP-1, and BCRP mRNA but not LRP mRNA assessed with RQ-PCR method in adult acute myeloid leukemia. Ann Hematol 2013; 93:577-93. [PMID: 24030729 DOI: 10.1007/s00277-013-1898-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/21/2013] [Accepted: 08/30/2013] [Indexed: 12/20/2022]
Abstract
Fms-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) and mixed-lineage leukemia gene-partial tandem duplication (MLL-PTD) are aberrations associated with leukemia which indicate unsatisfactory prognosis. Downstream regulatory targets of FLT3-ITD and MLL-PTD are not well defined. We have analyzed the expression of MDR-1, multidrug resistant protein-1 (MRP-1), breast cancer resistance protein (BCRP), and lung resistance protein (LRP) messenger RNA (mRNA) in relation to the mutational status of FLT3-ITD and MLL-PTD in 185 acute myeloid leukemia (AML) adult patients. The real-time quantitative polymerase chain reaction method was performed to assess the expression of the MDR-1, MRP-1, BCRP, and LRP mRNA, and the results were presented as coefficients calculated using an intermediate method according to Pfaffl's rule. Significantly higher expressions of MDR-1 mRNA were found in patients who did not harbor FLT3-ITD (0.20 vs. 0.05; p = 0.0001) and MRP-1 mRNA in patients with this mutation (0.96 vs. 0.70; p = 0.002) and of BCRP mRNA in patients with MLL-PTD (0.61 vs. 0.38; p = 0.03). In univariate analysis, the high expression of MDR-1 mRNA (≥0.1317) negatively influenced the outcome of induction therapy (p = 0.05), whereas the high expression of BCRP mRNA (≥1.1487) was associated with a high relapse rate (RR) (p = 0.013). We found that the high expression of MDR-1 (≥0.1317), MRP-1 (≥0.8409), and BCRP mRNA (≥1.1487) significantly influenced disease-free survival (DFS; p = 0.059, 0.032, and 0.009, respectively) and overall survival (0.048, 0.014, and 0.059, respectively). Moreover, a high expression of BCRP mRNA (≥1.1487) proved to be an independent prognostic factor for RR (p = 0.01) and DFS (p = 0.002) in multivariate analysis. The significant correlation between the expression of MDR-1, MRP-1, and BCRP mRNA and FLT3-ITD or MLL-PTD in AML patients requires further investigation.
Collapse
|
41
|
de Moraes ACR, Maranho CK, Rauber GS, Santos-Silva MC. Importance of detecting multidrug resistance proteins in acute leukemia prognosis and therapy. J Clin Lab Anal 2013; 27:62-71. [PMID: 23292860 PMCID: PMC6807608 DOI: 10.1002/jcla.21563] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/02/2012] [Accepted: 11/01/2012] [Indexed: 01/11/2023] Open
Abstract
Multidrug resistance (MDR) is a multifactorial phenomenon and the role of these proteins in generating the MDR phenotype is controversial. With this in mind, this review compiled the current data on the role of ABCB1, ABCC1, and LRP proteins in the prognosis of hematologic neoplasms and their influence on the choice of therapy. Literature showed that the detection of these proteins, mainly ABCB1, is important in the AL prognosis. However, there is controversy regarding the methodology used for their detection. In summary, the expression and activity profiles of ABCB1, ABCC1, and LRP, proteins capable of promoting the efflux of a variety of chemotherapeutic agents from the cell cytoplasm represent one of the greatest causes of failure in AL treatment.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/analysis
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Acute Disease
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Humans
- Leukemia/metabolism
- Leukemia/pathology
- Models, Biological
- Multidrug Resistance-Associated Proteins/analysis
- Multidrug Resistance-Associated Proteins/metabolism
- Prognosis
- Vault Ribonucleoprotein Particles/analysis
- Vault Ribonucleoprotein Particles/metabolism
Collapse
Affiliation(s)
- Ana Carolina Rabello de Moraes
- Programa de Pós-graduação em Farmácia, Universidade Federal de Santa Catarina, Campus Trindade, Florianópolis - SC, Brazil
| | | | | | | |
Collapse
|
42
|
Nakano A, Miki H, Nakamura S, Harada T, Oda A, Amou H, Fujii S, Kagawa K, Takeuchi K, Ozaki S, Matsumoto T, Abe M. Up-regulation of hexokinaseII in myeloma cells: targeting myeloma cells with 3-bromopyruvate. J Bioenerg Biomembr 2012; 44:31-8. [PMID: 22298254 DOI: 10.1007/s10863-012-9412-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/27/2011] [Accepted: 01/11/2012] [Indexed: 12/17/2022]
Abstract
Hexokinase II (HKII), a key enzyme of glycolysis, is widely over-expressed in cancer cells. However, HKII levels and its roles in ATP production and ATP-dependent cellular process have not been well studied in hematopoietic malignant cells including multiple myeloma (MM) cells.We demonstrate herein that HKII is constitutively over-expressed in MM cells. 3-bromopyruvate (3BrPA), an inhibitor of HKII, promptly and substantially suppresses ATP production and induces cell death in MM cells. Interestingly, cocultures with osteoclasts (OCs) but not bone marrow stromal cells (BMSCs) enhanced the phosphorylation of Akt along with an increase in HKII levels and lactate production in MM cells. The enhancement of HKII levels and lactate production in MM cells by OCs were mostly abrogated by the PI3K inhibitor LY294002, suggesting activation of glycolysis in MM cells by OCs via the PI3K-Akt-HKII pathway. Although BMSCs and OCs stimulate MM cell growth and survival, 3BrPA induces cell death in MM cells even in cocultures with OCs as well as BMSCs. Furthermore, 3BrPA was able to diminish ATP-dependent ABC transporter activity to restore drug retention in MM cells in the presence of OCs. These results may underpin possible clinical application of 3BrPA in patients with MM.
Collapse
Affiliation(s)
- Ayako Nakano
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Medicine, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yang J, Nagasawa DT, Spasic M, Amolis M, Choy W, Garcia HM, Prins RM, Liau LM, Yang I. Endogenous Vaults and Bioengineered Vault Nanoparticles for Treatment of Glioblastomas. Neurosurg Clin N Am 2012; 23:451-8. [DOI: 10.1016/j.nec.2012.04.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022]
|
44
|
Kim HP, Bernard L, Berkowitz J, Nitta J, Hogge DE. Flow cytometry-based assessment of mitoxantrone efflux from leukemic blasts varies with response to induction chemotherapy in acute myeloid leukemia. CYTOMETRY PART B-CLINICAL CYTOMETRY 2012; 82:283-94. [PMID: 22508650 DOI: 10.1002/cyto.b.21028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 10/12/2011] [Revised: 03/10/2012] [Accepted: 04/06/2012] [Indexed: 01/13/2023]
Abstract
BACKGROUND Accurate prediction of chemotherapy drug resistance would aid treatment decisions in acute myeloid leukemia (AML). The aim of this study was to determine if mitoxantrone efflux from AML blasts would correlate with response to induction chemotherapy. METHODS Flow cytometry was used to measure the median fluorescence intensity (MFI) for AML blasts incubated with mitoxantrone [an ATP-binding cassette (ABC) transporter substrate] with or without coincubation with cyclosporine A (a broad-spectrum inhibitor of ABC transporters) and a ratio (MFIR) between the inhibited and uninhibited MFI was calculated. RESULTS Among 174 AML patient blast samples, the mean MFIR for complete remission (CR) patients was lower than that obtained for induction failure (IF) patients (mean MFIR ± SD 1.62 ± 0.53 for CR after one cycle of chemotherapy vs. 2.22 ± 1.29 for CR after two cycles and 2.59 ± 0.98 for IF, P < 0.001). Logistic regression analysis determined 2.45 as the MFIR threshold above which 29% of patients achieved CR vs. a CR rate of 84% when the MFIR was ≤ 2.45 (P < 0.0001). In AML patients with normal karyotype (n = 80), CR was obtained for 33% of patients with an MFIR > 2.45 vs. 89% of those with MFIR ≤ 2.45 (P < 0.0001). In patients > age 60 (n = 77), 30% vs. 87% of those with MFIR > vs. ≤ 2.45 achieved CR (P < 0.0001). CONCLUSIONS This assay of ABC transporter function can potentially predict response to induction chemotherapy in AML.
Collapse
Affiliation(s)
- Hyun Pyo Kim
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | | | | | | | | |
Collapse
|
45
|
Natarajan K, Xie Y, Baer MR, Ross DD. Role of breast cancer resistance protein (BCRP/ABCG2) in cancer drug resistance. Biochem Pharmacol 2012; 83:1084-103. [PMID: 22248732 PMCID: PMC3307098 DOI: 10.1016/j.bcp.2012.01.002] [Citation(s) in RCA: 303] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/18/2011] [Revised: 01/02/2012] [Accepted: 01/03/2012] [Indexed: 01/16/2023]
Abstract
Since cloning of the ATP-binding cassette (ABC) family member breast cancer resistance protein (BCRP/ABCG2) and its characterization as a multidrug resistance efflux transporter in 1998, BCRP has been the subject of more than two thousand scholarly articles. In normal tissues, BCRP functions as a defense mechanism against toxins and xenobiotics, with expression in the gut, bile canaliculi, placenta, blood-testis and blood-brain barriers facilitating excretion and limiting absorption of potentially toxic substrate molecules, including many cancer chemotherapeutic drugs. BCRP also plays a key role in heme and folate homeostasis, which may help normal cells survive under conditions of hypoxia. BCRP expression appears to be a characteristic of certain normal tissue stem cells termed "side population cells," which are identified on flow cytometric analysis by their ability to exclude Hoechst 33342, a BCRP substrate fluorescent dye. Hence, BCRP expression may contribute to the natural resistance and longevity of these normal stem cells. Malignant tissues can exploit the properties of BCRP to survive hypoxia and to evade exposure to chemotherapeutic drugs. Evidence is mounting that many cancers display subpopulations of stem cells that are responsible for tumor self-renewal. Such stem cells frequently manifest the "side population" phenotype characterized by expression of BCRP and other ABC transporters. Along with other factors, these transporters may contribute to the inherent resistance of these neoplasms and their failure to be cured.
Collapse
Affiliation(s)
| | - Yi Xie
- University of Maryland Greenebaum Cancer Center
| | - Maria R. Baer
- University of Maryland Greenebaum Cancer Center
- Department of Medicine, University of Maryland School of Medicine
| | - Douglas D. Ross
- University of Maryland Greenebaum Cancer Center
- Department of Medicine, University of Maryland School of Medicine
- Departments of Pathology, and Pharmacology & Experimental Therapeutics, University of Maryland, School of Medicine
- Staff Physician, Baltimore VA Medical Center
| |
Collapse
|
46
|
She M, Niu X, Chen X, Li J, Zhou M, He Y, Le Y, Guo K. Resistance of leukemic stem-like cells in AML cell line KG1a to natural killer cell-mediated cytotoxicity. Cancer Lett 2011; 318:173-9. [PMID: 22198207 DOI: 10.1016/j.canlet.2011.12.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/07/2010] [Revised: 11/07/2011] [Accepted: 12/07/2011] [Indexed: 02/06/2023]
Abstract
Leukemic stem cells (LSCs) play the central role in the relapse and refractory of acute myeloid leukemia (AML) and highlight the critical need for the new therapeutic strategies to directly target the LSC population. However, relatively little is known about the unique molecular mechanisms of drug and natural killer cells (NK)-killing resistance of LSCs because of very small number of LSCs in bone marrow. In this study, we investigated whether established leukemia cell line contains LSCs. We showed that KG1a leukemia cell line contained leukemic stem-like cells, which have been phenotypically restricted within the CD34(+)CD38(-) fractions. CD34(+)CD38(-) cells could generate CD34(+)CD38(+) cells in culture medium and had renewal function. Moreover, CD34(+)CD38(-) cells had self-renewal potential. We found that leukemic stem-like cells from KG1a cells were resistant to chemotherapy and NK-mediated cytotoxicity. NKG2D ligands involve in protecting LSCs from NK-mediated attack. Taken together, our studies provide a novel cell model for leukemic stem cells research. Our data also shed light on mechanism of double resistant to chemotherapy and NK cell immunotherapy, which was helpful for developing novel effective strategies for LSCs.
Collapse
Affiliation(s)
- Miaorong She
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Nakanishi T, Ross DD. Breast cancer resistance protein (BCRP/ABCG2): its role in multidrug resistance and regulation of its gene expression. CHINESE JOURNAL OF CANCER 2011; 31:73-99. [PMID: 22098950 PMCID: PMC3777471 DOI: 10.5732/cjc.011.10320] [Citation(s) in RCA: 209] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022]
Abstract
Breast cancer resistance protein (BCRP)/ATP-binding cassette subfamily G member 2 (ABCG2) is an ATP-binding cassette (ABC) transporter identified as a molecular cause of multidrug resistance (MDR) in diverse cancer cells. BCRP physiologically functions as a part of a self-defense mechanism for the organism; it enhances elimination of toxic xenobiotic substances and harmful agents in the gut and biliary tract, as well as through the blood-brain, placental, and possibly blood-testis barriers. BCRP recognizes and transports numerous anticancer drugs including conventional chemotherapeutic and targeted small therapeutic molecules relatively new in clinical use. Thus, BCRP expression in cancer cells directly causes MDR by active efflux of anticancer drugs. Because BCRP is also known to be a stem cell marker, its expression in cancer cells could be a manifestation of metabolic and signaling pathways that confer multiple mechanisms of drug resistance, self-renewal (sternness), and invasiveness (aggressiveness), and thereby impart a poor prognosis. Therefore, blocking BCRP-mediated active efflux may provide a therapeutic benefit for cancers. Delineating the precise molecular mechanisms for BCRP gene expression may lead to identification of a novel molecular target to modulate BCRP-mediated MDR. Current evidence suggests that BCRP gene transcription is regulated by a number of trans-acting elements including hypoxia inducible factor 1α, estrogen receptor, and peroxisome proliferator-activated receptor. Furthermore, alternative promoter usage, demethylation of the BCRP promoter, and histone modification are likely associated with drug-induced BCRP overexpression in cancer cells. Finally, PI3K/AKT signaling may play a critical role in modulating BCRP function under a variety of conditions. These biological events seem involved in a complicated manner. Untangling the events would be an essential first step to developing a method to modulate BCRP function to aid patients with cancer. This review will present a synopsis of the impact of BCRP-mediated MDR in cancer cells, and the molecular mechanisms of acquired MDR currently postulated in a variety of human cancers.
Collapse
Affiliation(s)
- Takeo Nakanishi
- Department of Membrane Transport and Biopharmaceutics, Kanazawa University School of Pharmaceutical Sciences, Kanazawa, Japan.
| | | |
Collapse
|
48
|
Robey RW, Ierano C, Zhan Z, Bates SE. The challenge of exploiting ABCG2 in the clinic. Curr Pharm Biotechnol 2011; 12:595-608. [PMID: 21118093 DOI: 10.2174/138920111795163913] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/26/2010] [Accepted: 04/15/2010] [Indexed: 01/16/2023]
Abstract
ABCG2, or breast cancer resistance protein (BCRP), is an ATP-binding cassette half transporter that has been shown to transport a wide range of substrates including chemotherapeutics, antivirals, antibiotics and flavonoids. Given its wide range of substrates, much work has been dedicated to developing ABCG2 as a clinical target. But where can we intervene clinically and how can we avoid the mistakes made in past clinical trials targeting P-glycoprotein? This review will summarize the normal tissue distribution, cancer tissue expression, substrates and inhibitors of ABCG2, and highlight the challenges presented in exploiting ABCG2 in the clinic. We discuss the possibility of inhibiting ABCG2, so as to increase oral bioavailability or increase drug penetration into sanctuary sites, especially the central nervous system; and at the other end of the spectrum, the possibility of improving ABCG2 function, in the case of gout caused by a single nucleotide polymphism. Together, these aspects of ABCG2/BCRP make the protein a target of continuing interest for oncologists, biologists, and pharmacologists.
Collapse
Affiliation(s)
- Robert W Robey
- Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
49
|
Alvares CL, Schenk T, Hulkki S, Min T, Vijayaraghavan G, Yeung J, Gonzalez D, So CWE, Greaves M, Titley I, Bartolovic K, Morgan G. Tyrosine kinase inhibitor insensitivity of non-cycling CD34+ human acute myeloid leukaemia cells with FMS-like tyrosine kinase 3 mutations. Br J Haematol 2011; 154:457-65. [DOI: 10.1111/j.1365-2141.2011.08748.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/30/2022]
|
50
|
Witte KE, Ahlers J, Schäfer I, André M, Kerst G, Scheel-Walter HG, Schwarze CP, Pfeiffer M, Lang P, Handgretinger R, Ebinger M. High proportion of leukemic stem cells at diagnosis is correlated with unfavorable prognosis in childhood acute myeloid leukemia. Pediatr Hematol Oncol 2011; 28:91-9. [PMID: 21214408 DOI: 10.3109/08880018.2010.528171] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/14/2023]
Abstract
In acute myeloid leukemia (AML), the leukemia-initiating cell is found within the CD34(+)/CD38(-) cell compartment. Over the last years evidence grew that AML is initiated and propagated by leukemic stem cells (LSCs). Conceivably, these most immature leukemia cells are more resistant to therapy and subsequently initiate relapse. The authors studied 17 patients with childhood AML treated according to the AML-BFM 98/04 protocol. At diagnosis, the authors determined the characteristic immunophenotype of the leukemic cells by flow cytometry and investigated the expression of CD34, CD38, and CD45 to define a population of immunophenotypically immature cells (CD34(+)/CD38(-)/CD45(-/low)) enriched for LSCs in many cases of AML. The authors compared the fraction of this population of all myeloid cells at diagnosis with event-free survival. Kaplan-Meier analysis revealed significant higher event free survival of patients with low CD34(+)/CD38(-)/CD45(-/low) cell proportion (<0.68%) compared to patients with high burden of this population (>0.83%; log-rank P < .04). This correlation was not found for the total number of CD34(+) cells. This is the first study to show that a higher proportion of immature CD34(+)/CD38(-)/CD45(-/low) blasts at diagnosis correlates with unfavorable prognosis in childhood AML. The results suggest that a large CD34(+)/CD38(-)/CD45(-/low) population reflects a higher fraction of LSCs, leading to increased chemotherapy resistance and elevated relapse rate. Thus the initial frequency of CD34(+)/CD38(-)/CD45(-/low) cells may serve as a prognostic marker in pediatric AML. Future treatment in childhood AML should specifically target this immature population as well as the mature blast population.
Collapse
Affiliation(s)
- Kai-Erik Witte
- Department of Hematology/Oncology, University Children's Hospital, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|