1
|
Tian Y, Guo J, Mao L, Chen Z, Zhang X, Li Y, Zhang Y, Zha X, Luo OJ. Single-cell dissection reveals promotive role of ENO1 in leukemia stem cell self-renewal and chemoresistance in acute myeloid leukemia. Stem Cell Res Ther 2024; 15:347. [PMID: 39380054 PMCID: PMC11463110 DOI: 10.1186/s13287-024-03969-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Quiescent self-renewal of leukemia stem cells (LSCs) and resistance to conventional chemotherapy are the main factors leading to relapse of acute myeloid leukemia (AML). Alpha-enolase (ENO1), a key glycolytic enzyme, has been shown to regulate embryonic stem cell differentiation and promote self-renewal and malignant phenotypes in various cancer stem cells. Here, we sought to test whether and how ENO1 influences LSCs renewal and chemoresistance within the context of AML. METHODS We analyzed single-cell RNA sequencing data from bone marrow samples of 8 relapsed/refractory AML patients and 4 healthy controls using bioinformatics and machine learning algorithms. In addition, we compared ENO1 expression levels in the AML cohort with those in 37 control subjects and conducted survival analyses to correlate ENO1 expression with clinical outcomes. Furthermore, we performed functional studies involving ENO1 knockdown and inhibition in AML cell line. RESULTS We used machine learning to model and infer malignant cells in AML, finding more primitive malignant cells in the non-response (NR) group. The differentiation capacity of LSCs and progenitor malignant cells exhibited an inverse correlation with glycolysis levels. Trajectory analysis indicated delayed myeloid cell differentiation in NR group, with high ENO1-expressing LSCs at the initial stages of differentiation being preserved post-treatment. Simultaneously, ENO1 and stemness-related genes were upregulated and co-expressed in malignant cells during early differentiation. ENO1 level in our AML cohort was significantly higher than the controls, with higher levels in NR compared to those in complete remission. Knockdown of ENO1 in AML cell line resulted in the activation of LSCs, promoting cell differentiation and apoptosis, and inhibited proliferation. ENO1 inhibitor can impede the proliferation of AML cells. Furthermore, survival analyses associated higher ENO1 expression with poorer outcome in AML patients. CONCLUSIONS Our findings underscore the critical role of ENO1 as a plausible driver of LSC self-renewal, a potential target for AML target therapy and a biomarker for AML prognosis.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Phosphopyruvate Hydratase/metabolism
- Phosphopyruvate Hydratase/genetics
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Tumor Suppressor Proteins/metabolism
- Tumor Suppressor Proteins/genetics
- Female
- Drug Resistance, Neoplasm
- Single-Cell Analysis
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/genetics
- Male
- Middle Aged
- Cell Self Renewal
- Adult
- Cell Line, Tumor
- Cell Differentiation
- Aged
- Biomarkers, Tumor
Collapse
Affiliation(s)
- Yun Tian
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jiafan Guo
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Lipeng Mao
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhixi Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Xingwei Zhang
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China.
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Yikai Zhang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China.
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China.
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
2
|
El Khawanky N, Hughes A, Yu W, Myburgh R, Matschulla T, Taromi S, Aumann K, Clarson J, Vinnakota JM, Shoumariyeh K, Miething C, Lopez AF, Brown MP, Duyster J, Hein L, Manz MG, Hughes TP, White DL, Yong ASM, Zeiser R. Demethylating therapy increases anti-CD123 CAR T cell cytotoxicity against acute myeloid leukemia. Nat Commun 2021; 12:6436. [PMID: 34750374 PMCID: PMC8575966 DOI: 10.1038/s41467-021-26683-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/19/2021] [Indexed: 12/18/2022] Open
Abstract
Successful treatment of acute myeloid leukemia (AML) with chimeric antigen receptor (CAR) T cells is hampered by toxicity on normal hematopoietic progenitor cells and low CAR T cell persistence. Here, we develop third-generation anti-CD123 CAR T cells with a humanized CSL362-based ScFv and a CD28-OX40-CD3ζ intracellular signaling domain. This CAR demonstrates anti-AML activity without affecting the healthy hematopoietic system, or causing epithelial tissue damage in a xenograft model. CD123 expression on leukemia cells increases upon 5'-Azacitidine (AZA) treatment. AZA treatment of leukemia-bearing mice causes an increase in CTLA-4negative anti-CD123 CAR T cell numbers following infusion. Functionally, the CTLA-4negative anti-CD123 CAR T cells exhibit superior cytotoxicity against AML cells, accompanied by higher TNFα production and enhanced downstream phosphorylation of key T cell activation molecules. Our findings indicate that AZA increases the immunogenicity of AML cells, enhancing recognition and elimination of malignant cells by highly efficient CTLA-4negative anti-CD123 CAR T cells.
Collapse
MESH Headings
- Acute Disease
- Animals
- Azacitidine/administration & dosage
- Cell Line, Tumor
- Cells, Cultured
- Cytotoxicity, Immunologic
- DNA Methylation/drug effects
- Enzyme Inhibitors/administration & dosage
- HEK293 Cells
- HL-60 Cells
- Humans
- Immunotherapy, Adoptive/methods
- Interleukin-3 Receptor alpha Subunit/immunology
- Interleukin-3 Receptor alpha Subunit/metabolism
- Kaplan-Meier Estimate
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/pathology
- Leukemia, Myeloid/therapy
- Mice, Knockout
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Single-Chain Antibodies/immunology
- Xenograft Model Antitumor Assays/methods
- Mice
Collapse
Affiliation(s)
- Nadia El Khawanky
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Amy Hughes
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Wenbo Yu
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Renier Myburgh
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Comprehensive Cancer Center Zurich (CCCZ), Zurich, Switzerland
| | - Tony Matschulla
- Institute of Experimental and Clinical Pharmacology and Toxicology, Division II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sanaz Taromi
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Medical and Life Sciences, University Furtwangen, Villingen-Schwenningen, Germany
| | - Konrad Aumann
- Department of Pathology, Institute for Clinical Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Jade Clarson
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Janaki Manoja Vinnakota
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Khalid Shoumariyeh
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius Miething
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angel F Lopez
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Michael P Brown
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Cancer Clinical Trials Unit, Department of Medical Oncology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Justus Duyster
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Division II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Comprehensive Cancer Center Zurich (CCCZ), Zurich, Switzerland
| | - Timothy P Hughes
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Deborah L White
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- School of Biological Sciences, Faculty of Science, University of Adelaide, Adelaide, SA, Australia
| | - Agnes S M Yong
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.
- Department of Haematology, Royal Perth Hospital, Perth, WA, Australia.
- School of Medicine, The University of Western Australia, Perth, WA, Australia.
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signaling Research Centres BIOSS and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Jones CL, Inguva A, Jordan CT. Targeting Energy Metabolism in Cancer Stem Cells: Progress and Challenges in Leukemia and Solid Tumors. Cell Stem Cell 2021; 28:378-393. [PMID: 33667359 PMCID: PMC7951949 DOI: 10.1016/j.stem.2021.02.013] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malignant stem cells have long been considered a key therapeutic target in leukemia. Therapeutic strategies designed to target the fundamental biology of leukemia stem cells while sparing normal hematopoietic cells may provide better outcomes for leukemia patients. One process in leukemia stem cell biology that has intriguing therapeutic potential is energy metabolism. In this article we discuss the metabolic properties of leukemia stem cells and how targeting energy metabolism may provide more effective therapeutic regimens for leukemia patients. In addition, we highlight the similarities and differences in energy metabolism between leukemia stem cells and malignant stem cells from solid tumors.
Collapse
Affiliation(s)
- Courtney L Jones
- Princess Margaret Cancer Centre, 101 College St. Toronto, ON M5G 1L7, Canada
| | - Anagha Inguva
- Division of Hematology, University of Colorado, 12700 East 19th Ave., Aurora, CO 80045, USA
| | - Craig T Jordan
- Division of Hematology, University of Colorado, 12700 East 19th Ave., Aurora, CO 80045, USA.
| |
Collapse
|
4
|
Park CS, Lewis AH, Chen TJ, Bridges CS, Shen Y, Suppipat K, Puppi M, Tomolonis JA, Pang PD, Mistretta TA, Ma L, Green MR, Rau R, Lacorazza HD. A KLF4-DYRK2-mediated pathway regulating self-renewal in CML stem cells. Blood 2019; 134:1960-1972. [PMID: 31515251 PMCID: PMC6887114 DOI: 10.1182/blood.2018875922] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/06/2019] [Indexed: 02/02/2023] Open
Abstract
Leukemia stem cells are a rare population with a primitive progenitor phenotype that can initiate, sustain, and recapitulate leukemia through a poorly understood mechanism of self-renewal. Here, we report that Krüppel-like factor 4 (KLF4) promotes disease progression in a murine model of chronic myeloid leukemia (CML)-like myeloproliferative neoplasia by repressing an inhibitory mechanism of preservation in leukemia stem/progenitor cells with leukemia-initiating capacity. Deletion of the Klf4 gene severely abrogated the maintenance of BCR-ABL1(p210)-induced CML by impairing survival and self-renewal in BCR-ABL1+ CD150+ lineage-negative Sca-1+ c-Kit+ leukemic cells. Mechanistically, KLF4 repressed the Dyrk2 gene in leukemic stem/progenitor cells; thus, loss of KLF4 resulted in elevated levels of dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 2 (DYRK2), which were associated with inhibition of survival and self-renewal via depletion of c-Myc protein and p53 activation. In addition to transcriptional regulation, stabilization of DYRK2 protein by inhibiting ubiquitin E3 ligase SIAH2 with vitamin K3 promoted apoptosis and abrogated self-renewal in murine and human CML stem/progenitor cells. Altogether, our results suggest that DYRK2 is a molecular checkpoint controlling p53- and c-Myc-mediated regulation of survival and self-renewal in CML cells with leukemic-initiating capacity that can be targeted with small molecules.
Collapse
MESH Headings
- Animals
- Cell Survival/drug effects
- Cell Survival/genetics
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Gene Deletion
- Humans
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mice, Knockout
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Signal Transduction
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
- Vitamin K 3/pharmacology
- Dyrk Kinases
Collapse
Affiliation(s)
| | - Andrew H Lewis
- Department of Pathology and Immunology and
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX
| | - Taylor J Chen
- Department of Pathology and Immunology and
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX
| | | | - Ye Shen
- Department of Pathology and Immunology and
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX
| | - Koramit Suppipat
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Houston, TX
| | | | | | - Paul D Pang
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX
| | | | - Leyuan Ma
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Michael R Green
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Rachel Rau
- Department of Pediatrics-Oncology, Baylor College of Medicine, Houston, TX
| | | |
Collapse
|
5
|
Fan M, Li M, Gao L, Geng S, Wang J, Wang Y, Yan Z, Yu L. Chimeric antigen receptors for adoptive T cell therapy in acute myeloid leukemia. J Hematol Oncol 2017; 10:151. [PMID: 28851445 PMCID: PMC5576380 DOI: 10.1186/s13045-017-0519-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/17/2017] [Indexed: 12/17/2022] Open
Abstract
Currently, conventional therapies for acute myeloid leukemia (AML) have high failure and relapse rates. Thus, developing new strategies is crucial for improving the treatment of AML. With the clinical success of anti-CD19 chimeric antigen receptor (CAR) T cell therapies against B-lineage malignancies, many studies have attempted to translate the success of CAR T cell therapy to other malignancies, including AML. This review summarizes the current advances in CAR T cell therapy against AML, including preclinical studies and clinical trials, and discusses the potential AML-associated surface markers that could be used for further CAR technology. Finally, we describe strategies that might address the current issues of employing CAR T cell therapy in AML.
Collapse
Affiliation(s)
- Mingxue Fan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO. 3663 Zhongshan Road, Shanghai, 200062, People's Republic of China
| | - Minghao Li
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO. 3663 Zhongshan Road, Shanghai, 200062, People's Republic of China
| | - Lipeng Gao
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO. 3663 Zhongshan Road, Shanghai, 200062, People's Republic of China
| | - Sicong Geng
- China Novartis Institutes for Biomedical Research Co., Ltd., GDD/TRD/Chemical and Pharmaceutical Profiling, 5F, Building 3, Novartis Campus 4218 Jinke Rd, Zhangjiang Hi-Tech Park Pudong District, Shanghai, 201203, China
| | - Jing Wang
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO. 3663 Zhongshan Road, Shanghai, 200062, People's Republic of China
| | - Yiting Wang
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO. 3663 Zhongshan Road, Shanghai, 200062, People's Republic of China
| | - Zhiqiang Yan
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO. 3663 Zhongshan Road, Shanghai, 200062, People's Republic of China.
| | - Lei Yu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO. 3663 Zhongshan Road, Shanghai, 200062, People's Republic of China.
| |
Collapse
|
6
|
Xie LH, Biondo M, Busfield SJ, Arruda A, Yang X, Vairo G, Minden MD. CD123 target validation and preclinical evaluation of ADCC activity of anti-CD123 antibody CSL362 in combination with NKs from AML patients in remission. Blood Cancer J 2017; 7:e567. [PMID: 28574487 PMCID: PMC5520399 DOI: 10.1038/bcj.2017.52] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/24/2017] [Indexed: 01/05/2023] Open
Abstract
Despite the heterogeneity of acute myeloid leukemia (AML), overexpression of the interleukin-3 receptor-α (CD123) on both the more differentiated leukemic blast and leukemic stem cells (LSCs) provides a therapeutic target for antibody treatment. Here we present data on the potential clinical activity of the monoclonal antibody CSL362, which binds to CD123 with high affinity. We first validated the expression of CD123 by 100% (52/52) of patient samples and the correlation of NPM1 and FLT3-ITD mutations with the high frequency of CD123 in AML. In vitro studies demonstrated that CSL362 potently induced antibody-dependent cell cytotoxicity (ADCC) of AML blasts including CD34+CD38−CD123+ LSCs by natural killer cells (NKs). Importantly, compared with healthy donor (HD) NKs, NKs drawn from AML patients in remission had a comparable ADCC activity against leukemic cells; of note, during remission, immature NKs were five times higher in AML patients than that in HDs. Significantly, we report a case where leukemic cells were resistant to autologous ADCC; however, the blasts were effectively lysed by CSL362 together with donor-derived NKs after allogeneic hematopoietic stem cell transplantation. These studies highlight CSL362 as a promising therapeutic option following chemotherapy and transplant so as to improve the outcome of AML patients.
Collapse
Affiliation(s)
- L H Xie
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - M Biondo
- CSL Limited, Bio21 Institute, Parkville, Victoria, Australia
| | - S J Busfield
- CSL Limited, Bio21 Institute, Parkville, Victoria, Australia
| | - A Arruda
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - X Yang
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - G Vairo
- CSL Limited, Bio21 Institute, Parkville, Victoria, Australia
| | - M D Minden
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Pei S, Minhajuddin M, D'Alessandro A, Nemkov T, Stevens BM, Adane B, Khan N, Hagen FK, Yadav VK, De S, Ashton JM, Hansen KC, Gutman JA, Pollyea DA, Crooks PA, Smith C, Jordan CT. Rational Design of a Parthenolide-based Drug Regimen That Selectively Eradicates Acute Myelogenous Leukemia Stem Cells. J Biol Chem 2016; 291:21984-22000. [PMID: 27573247 DOI: 10.1074/jbc.m116.750653] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Indexed: 12/22/2022] Open
Abstract
Although multidrug approaches to cancer therapy are common, few strategies are based on rigorous scientific principles. Rather, drug combinations are largely dictated by empirical or clinical parameters. In the present study we developed a strategy for rational design of a regimen that selectively targets human acute myelogenous leukemia (AML) stem cells. As a starting point, we used parthenolide, an agent shown to target critical mechanisms of redox balance in primary AML cells. Next, using proteomic, genomic, and metabolomic methods, we determined that treatment with parthenolide leads to induction of compensatory mechanisms that include up-regulated NADPH production via the pentose phosphate pathway as well as activation of the Nrf2-mediated oxidative stress response pathway. Using this knowledge we identified 2-deoxyglucose and temsirolimus as agents that can be added to a parthenolide regimen as a means to inhibit such compensatory events and thereby further enhance eradication of AML cells. We demonstrate that the parthenolide, 2-deoxyglucose, temsirolimus (termed PDT) regimen is a potent means of targeting AML stem cells but has little to no effect on normal stem cells. Taken together our findings illustrate a comprehensive approach to designing combination anticancer drug regimens.
Collapse
Affiliation(s)
| | | | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado 80045
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado 80045
| | | | | | | | | | - Vinod K Yadav
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, and
| | - Subhajyoti De
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, and
| | - John M Ashton
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York 14642
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado 80045
| | | | | | - Peter A Crooks
- Department of Pharmaceutical Sciences, University of Arkansas, Little Rock, Arkansas 72205
| | | | | |
Collapse
|
8
|
Evolution of acute myelogenous leukemia stem cell properties after treatment and progression. Blood 2016; 128:1671-8. [PMID: 27421961 DOI: 10.1182/blood-2016-02-695312] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/28/2016] [Indexed: 12/12/2022] Open
Abstract
Most cancers evolve over time as patients initially responsive to therapy acquire resistance to the same drugs at relapse. Cancer stem cells have been postulated to represent a therapy-refractory reservoir for relapse, but formal proof of this model is lacking. We prospectively characterized leukemia stem cell populations (LSCs) from a well-defined cohort of patients with acute myelogenous leukemia (AML) at diagnosis and relapse to assess the effect of the disease course on these critical populations. Leukemic samples were collected from patients with newly diagnosed AML before therapy and after relapse, and LSC frequency was assessed by limiting dilution analyses. LSC populations were identified using fluorescent-labeled cell sorting and transplantation into immunodeficient NOD/SCID/interleukin 2 receptor γ chain null mice. The surface antigen expression profiles of pretherapy and postrelapse LSCs were determined for published LSC markers. We demonstrate a 9- to 90-fold increase in LSC frequency between diagnosis and relapse. LSC activity at relapse was identified in populations of leukemic blasts that did not demonstrate this activity before treatment and relapse. In addition, we describe genetic instability and exceptional phenotypic changes that accompany the evolution of these new LSC populations. This study is the first to characterize the evolution of LSCs in vivo after chemotherapy, identifying a dramatic change in the physiology of primitive AML cells when the disease progresses. Taken together, these findings provide a new frame of reference by which to evaluate candidate AML therapies in which both disease control and the induction of more advanced forms of disease should be considered.
Collapse
|
9
|
Daria D, Kirsten N, Muranyi A, Mulaw M, Ihme S, Kechter A, Hollnagel M, Bullinger L, Döhner K, Döhner H, Feuring-Buske M, Buske C. GPR56 contributes to the development of acute myeloid leukemia in mice. Leukemia 2016; 30:1734-41. [PMID: 27063597 DOI: 10.1038/leu.2016.76] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/02/2016] [Accepted: 03/14/2016] [Indexed: 12/19/2022]
Abstract
The G protein-coupled receptor 56 (GPR56) was identified as part of the molecular signature of functionally validated leukemic stem cells isolated from patients with acute myeloid leukemia (AML). This report now demonstrates particularly high expression of GPR56 in patients with mutant NPM1 and FLT3-length mutation and association of high GPR56 expression with inferior prognosis in a large patient cohort treated in two independent multicenter phase III trials. Functional relevance of GPR56 expression was validated in mice, in which co-expression of Gpr56 significantly accelerated HOXA9-induced leukemogenesis and vice versa knockdown of Gpr56 delayed onset of HOXA9/MEIS1-induced AML. Overexpression of Gpr56 grossly changed the molecular phenotype of Hoxa9-transduced cells affecting pathways involved in G protein-coupled receptors (GPRCs) and associated intracellular signaling. Blockage of surface GPR56 by an anti-GPR56 antibody successfully impaired engraftment of primary human AML cells. In summary, these data demonstrate that high expression of GPR56 is able to contribute to AML development and characterize the GPR56 as a potential novel target for antibody-mediated antileukemic strategies.
Collapse
Affiliation(s)
- D Daria
- Institute for Experimental Cancer Research, Comprehensive Cancer Center, University Hospital of Ulm, Ulm, Germany
| | - N Kirsten
- Institute for Experimental Cancer Research, Comprehensive Cancer Center, University Hospital of Ulm, Ulm, Germany
| | - A Muranyi
- Institute for Experimental Cancer Research, Comprehensive Cancer Center, University Hospital of Ulm, Ulm, Germany
| | - M Mulaw
- Institute for Experimental Cancer Research, Comprehensive Cancer Center, University Hospital of Ulm, Ulm, Germany
| | - S Ihme
- Institute for Experimental Cancer Research, Comprehensive Cancer Center, University Hospital of Ulm, Ulm, Germany
| | - A Kechter
- Institute for Experimental Cancer Research, Comprehensive Cancer Center, University Hospital of Ulm, Ulm, Germany
| | - M Hollnagel
- Institute for Experimental Cancer Research, Comprehensive Cancer Center, University Hospital of Ulm, Ulm, Germany
| | - L Bullinger
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - K Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - H Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - M Feuring-Buske
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - C Buske
- Institute for Experimental Cancer Research, Comprehensive Cancer Center, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
10
|
Gao XM, Zhang R, Dong QZ, Qin LX. Properties and feasibility of using cancer stem cells in clinical cancer treatment. Cancer Biol Med 2016; 13:489-495. [PMID: 28154781 PMCID: PMC5250607 DOI: 10.20892/j.issn.2095-3941.2016.0076] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer treatment failure, drug resistance, or metastatic recurrence are thought to be caused mainly by the existence of a very small number of cancer stem cells (CSCs). The characteristics of this subgroup of cells include self-renewal, tumorigenesis, multiple differentiation and high invasiveness, metastasis, and drug resistance potential. Many studies have demonstrated that CSCs play important roles in tumor growth, spread and metastatic relapse after treatment, and are closely related to the prognosis of patients. From a therapeutic viewpoint, deep insights into the CSCs biology, development of specific therapeutic strategies for targeting CSCs, and characterization of their microenvironment could be an ideal way to combat cancer.
Collapse
Affiliation(s)
- Xiao-Mei Gao
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Rui Zhang
- Department of General Surgery, Huashan Hospital and Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Qiong-Zhu Dong
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of General Surgery, Huashan Hospital and Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Lun-Xiu Qin
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of General Surgery, Huashan Hospital and Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| |
Collapse
|
11
|
Sugimori M, Hayakawa Y, Boman BM, Fields JZ, Awaji M, Kozano H, Tamura R, Yamamoto S, Ogata T, Yamada M, Endo S, Kurimoto M, Kuroda S. Discovery of Power-Law Growth in the Self-Renewal of Heterogeneous Glioma Stem Cell Populations. PLoS One 2015; 10:e0135760. [PMID: 26284929 PMCID: PMC4540573 DOI: 10.1371/journal.pone.0135760] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/24/2015] [Indexed: 01/06/2023] Open
Abstract
Background Accumulating evidence indicates that cancer stem cells (CSCs) drive tumorigenesis. This suggests that CSCs should make ideal therapeutic targets. However, because CSC populations in tumors appear heterogeneous, it remains unclear how CSCs might be effectively targeted. To investigate the mechanisms by which CSC populations maintain heterogeneity during self-renewal, we established a glioma sphere (GS) forming model, to generate a population in which glioma stem cells (GSCs) become enriched. We hypothesized, based on the clonal evolution concept, that with each passage in culture, heterogeneous clonal sublines of GSs are generated that progressively show increased proliferative ability. Methodology/Principal Findings To test this hypothesis, we determined whether, with each passage, glioma neurosphere culture generated from four different glioma cell lines become progressively proliferative (i.e., enriched in large spheres). Rather than monitoring self-renewal, we measured heterogeneity based on neurosphere clone sizes (#cells/clone). Log-log plots of distributions of clone sizes yielded a good fit (r>0.90) to a straight line (log(% total clones) = k*log(#cells/clone)) indicating that the system follows a power-law (y = xk) with a specific degree exponent (k = −1.42). Repeated passaging of the total GS population showed that the same power-law was maintained over six passages (CV = −1.01 to −1.17). Surprisingly, passage of either isolated small or large subclones generated fully heterogeneous populations that retained the original power-law-dependent heterogeneity. The anti-GSC agent Temozolomide, which is well known as a standard therapy for glioblastoma multiforme (GBM), suppressed the self-renewal of clones, but it never disrupted the power-law behavior of a GS population. Conclusions/Significance Although the data above did not support the stated hypothesis, they did strongly suggest a novel mechanism that underlies CSC heterogeneity. They indicate that power-law growth governs the self-renewal of heterogeneous glioma stem cell populations. That the data always fit a power-law suggests that: (i) clone sizes follow continuous, non-random, and scale-free hierarchy; (ii) precise biologic rules that reflect self-organizing emergent behaviors govern the generation of neurospheres. That the power-law behavior and the original GS heterogeneity are maintained over multiple passages indicates that these rules are invariant. These self-organizing mechanisms very likely underlie tumor heterogeneity during tumor growth. Discovery of this power-law behavior provides a mechanism that could be targeted in the development of new, more effective, anti-cancer agents.
Collapse
Affiliation(s)
- Michiya Sugimori
- Department of Integrative Neuroscience, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
- * E-mail:
| | - Yumiko Hayakawa
- Department of Neurosurgery, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Bruce M. Boman
- Center for Translational Cancer Research, Departments of Biology and Mathematics, University of Delaware, Helen F Graham Cancer Center and Research Institute, Newark, DE 19711 United States of America
| | - Jeremy Z. Fields
- Biotechnical Research, CATX, Inc., Gladwyne, PA 19035 United States of America
| | - Miharu Awaji
- Department of Integrative Neuroscience, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Hiroko Kozano
- Department of Integrative Neuroscience, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Ryoi Tamura
- Department of Integrative Neuroscience, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Seiji Yamamoto
- Department of Pathology, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Toru Ogata
- The Center of Sports Science and Health Promotion in the NRCD Hospital, National Rehabilitation Center for Persons with Disabilities, 4–1 Namiki, Tokorozawa, Saitama 359–8555, Japan
| | - Mitsuhiko Yamada
- Department of Neuropsycopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187–8553, Japan
| | - Shunro Endo
- Department of Neurosurgery, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Masanori Kurimoto
- Department of Neurosurgery, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| | - Satoshi Kuroda
- Department of Neurosurgery, University of Toyama, 2630 Sugitani, Toyama, Toyama 930–0194, Japan
| |
Collapse
|
12
|
Kempema AM, Widen JC, Hexum JK, Andrews TE, Wang D, Rathe SK, Meece FA, Noble KE, Sachs Z, Largaespada DA, Harki DA. Synthesis and antileukemic activities of C1-C10-modified parthenolide analogues. Bioorg Med Chem 2015; 23:4737-4745. [PMID: 26088334 DOI: 10.1016/j.bmc.2015.05.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 05/24/2015] [Indexed: 01/08/2023]
Abstract
Parthenolide (PTL) is a sesquiterpene lactone natural product with anti-proliferative activity to cancer cells. Selective eradication of leukemic stem cells (LSCs) over healthy hematopoietic stem cells (HSCs) by PTL has been demonstrated in previous studies, which suggests PTL and related molecules may be useful for targeting LSCs. Eradication of LSCs is required for curative therapy. Chemical optimizations of PTL to improve potency and pharmacokinetic parameters have focused largely on the α-methylene-γ-butyrolactone, which is essential for activity. Conversely, we evaluated modifications to the C1-C10 olefin and benchmarked new inhibitors to PTL with respect to inhibitory potency across a panel of cancer cell lines, ability to target drug-resistant acute myeloid leukemia (AML) cells, efficacy for inhibiting clonal growth of AML cells, toxicity to healthy bone marrow cells, and efficiency for promoting intracellular reactive oxygen species (ROS) levels. Cyclopropane 4 was found to possess less toxicity to healthy bone marrow cells, enhanced potency for the induction of cellular ROS, and similar broad-spectrum anti-proliferative activity to cancer cells in comparison to PTL.
Collapse
Affiliation(s)
- Aaron M Kempema
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - John C Widen
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph K Hexum
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Timothy E Andrews
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dan Wang
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Susan K Rathe
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Frederick A Meece
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Klara E Noble
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zohar Sachs
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - David A Largaespada
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
13
|
Rai S, Tanaka H, Suzuki M, Ogoh H, Taniguchi Y, Morita Y, Shimada T, Tanimura A, Matsui K, Yokota T, Oritani K, Tanabe K, Watanabe T, Kanakura Y, Matsumura I. Clathrin assembly protein CALM plays a critical role in KIT signaling by regulating its cellular transport from early to late endosomes in hematopoietic cells. PLoS One 2014; 9:e109441. [PMID: 25279552 PMCID: PMC4184852 DOI: 10.1371/journal.pone.0109441] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 09/07/2014] [Indexed: 11/29/2022] Open
Abstract
CALM is implicated in the formation of clathrin-coated vesicles, which mediate endocytosis and intracellular trafficking of growth factor receptors and nutrients. We previously found that CALM-deficient mice suffer from severe anemia due to the impaired clathrin-mediated endocytosis of transferrin receptor in immature erythroblast. However, CALM has been supposed to regulate the growth and survival of hematopoietic stem/progenitor cells. So, in this study, we focused on the function of CALM in these cells. We here show that the number of Linage−Sca-1+KIT+ (LSK) cells decreased in the fetal liver of CALM−/− mice. Also, colony forming activity was impaired in CALM−/− LSK cells. In addition, SCF, FLT3, and TPO-dependent growth was severely impaired in CALM−/− LSK cells, while they can normally proliferate in response to IL-3 and IL-6. We also examined the intracellular trafficking of KIT using CALM−/− murine embryonic fibroblasts (MEFs) engineered to express KIT. At first, we confirmed that endocytosis of SCF-bound KIT was not impaired in CALM−/− MEFs by the internalization assay. However, SCF-induced KIT trafficking from early to late endosome was severely impaired in CALM−/− MEFs. As a result, although intracellular KIT disappeared 30 min after SCF stimulation in wild-type (WT) MEFs, it was retained in CALM−/− MEFs. Furthermore, SCF-induced phosphorylation of cytosolic KIT was enhanced and prolonged in CALM−/− MEFs compared with that in WT MEFs, leading to the excessive activation of Akt. Similar hyperactivation of Akt was observed in CALM−/− KIT+ cells. These results indicate that CALM is essential for the intracellular trafficking of KIT and its normal functions. Also, our data demonstrate that KIT located in the early endosome can activate downstream molecules as a signaling endosome. Because KIT activation is involved in the pathogenesis of some malignancies, the manipulation of CALM function would be an attractive therapeutic strategy.
Collapse
Affiliation(s)
- Shinya Rai
- Department of Hematology and Rheumatology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Hirokazu Tanaka
- Department of Hematology and Rheumatology, Kinki University Faculty of Medicine, Osaka, Japan
- * E-mail:
| | - Mai Suzuki
- Division of Hematological Malignancy, National Cancer Center Research Institute, Tokyo, Japan
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women’s University, Nara, Japan
| | - Honami Ogoh
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women’s University, Nara, Japan
| | - Yasuhiro Taniguchi
- Department of Hematology and Rheumatology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Yasuyoshi Morita
- Department of Hematology and Rheumatology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Takahiro Shimada
- Department of Hematology and Rheumatology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Akira Tanimura
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keiko Matsui
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takafumi Yokota
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenji Oritani
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenji Tanabe
- Medical Research Institute, Tokyo Women’s Medical University, Tokyo, Japan
| | - Toshio Watanabe
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women’s University, Nara, Japan
| | - Yuzuru Kanakura
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kinki University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
14
|
Smith C, Gasparetto M, Humphries K, Pollyea DA, Vasiliou V, Jordan CT. Aldehyde dehydrogenases in acute myeloid leukemia. Ann N Y Acad Sci 2014; 1310:58-68. [PMID: 24641679 DOI: 10.1111/nyas.12414] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Acute myeloid leukemia (AML) affects approximately 15,000 persons per year in the United States and is the sixth leading cause of cancer-related deaths. The treatment of AML has advanced little in the past thirty years, in part because of the biologic heterogeneity of the disease and the difficulty in targeting AML cells while sparing normal hematopoietic cells. Advances in preventing and treating AML are likely to occur once the cellular and molecular differences between leukemia and normal hematopoietic cells are better understood. Aldehyde dehydrogenase (ALDH) activity is highly expressed in hematopoietic stem cells (HSCs), while, in contrast, a subset of AMLs are lacking this activity. This difference may be relevant to the development of AML and may also provide a better avenue for treating this disease. In this review, we summarize what is known about the ALDHs in normal HSCs and AML and propose strategies for capitalizing on these differences in the treatment of acute leukemia, and possibly other cancers as well.
Collapse
Affiliation(s)
- Clay Smith
- Division of Hematology, University of Colorado, Aurora, Colorado
| | | | | | | | | | | |
Collapse
|
15
|
Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood 2014; 123:2343-54. [PMID: 24596416 DOI: 10.1182/blood-2013-09-529537] [Citation(s) in RCA: 372] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Many patients with acute myeloid leukemia (AML) are incurable with chemotherapy and may benefit from novel approaches. One such approach involves the transfer of T cells engineered to express chimeric antigen receptors (CARs) for a specific cell-surface antigen. This strategy depends upon preferential expression of the target on tumor cells. To date, the lack of AML-specific surface markers has impeded development of such CAR-based approaches. CD123, the transmembrane α chain of the interleukin-3 receptor, is expressed in the majority of AML cells but is also expressed in many normal hematopoietic cells. Here, we show that CD123 is a good target for AML-directed CAR therapy, because its expression increases over time in vivo even in initially CD123(dim) populations, and that human CD123-redirected T cells (CART123) eradicate primary AML in immunodeficient mice. CART123 also eradicated normal human myelopoiesis, a surprising finding because anti-CD123 antibody-based strategies have been reportedly well tolerated. Because AML is likely preceded by clonal evolution in "preleukemic" hematopoietic stem cells, our observations support CART123 as a viable AML therapy, suggest that CART123-based myeloablation may be used as a novel conditioning regimen for hematopoietic cell transplantation, and raise concerns for the use of CART123 without such a rescue strategy.
Collapse
|
16
|
Braciak TA, Wildenhain S, Roskopf CC, Schubert IA, Fey GH, Jacob U, Hopfner KP, Oduncu FS. NK cells from an AML patient have recovered in remission and reached comparable cytolytic activity to that of a healthy monozygotic twin mediated by the single-chain triplebody SPM-2. J Transl Med 2013; 11:289. [PMID: 24237598 PMCID: PMC3842817 DOI: 10.1186/1479-5876-11-289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 11/14/2013] [Indexed: 12/26/2022] Open
Abstract
Background The capacity of patient’s Natural Killer cells (NKs) to be activated for cytolysis is an important prerequisite for the success of antibody-derived agents such as single-chain triplebodies (triplebodies) in cancer therapy. NKs recovered from AML patients at diagnosis are often found to be reduced in peripheral blood titers and cytolytic activity. Here, we had the unique opportunity to compare blood titers and cytolytic function of NKs from an AML patient with those of a healthy monozygotic twin. The sibling’s NKs were compared with the patient’s drawn either at diagnosis or in remission after chemotherapy. The cytolytic activities of NKs from these different sources for the patient’s autologous AML blasts and other leukemic target cells in conjunction with triplebody SPM-2, targeting the surface antigens CD33 and CD123 on the AML cells, were compared. Methods Patient NKs drawn at diagnosis were compared to NKs drawn in remission after chemotherapy and a sibling’s NKs, all prepared from PBMCs by immunomagnetic beads (MACS). Redirected lysis (RDL) assays using SPM-2 and antibody-dependent cellular cytotoxicity (ADCC) assays using the therapeutic antibody RituximabTM were performed with the enriched NKs. In addition, MACS-sorted NKs were analyzed for NK cell activating receptors (NCRs) by flow cytometry, and the release of TNF-alpha and IFN-gamma from blood samples of both siblings after the addition of the triplebody were measured in ELISA-assays. Results Patient NKs isolated from peripheral blood drawn in remission produced comparable lysis as NKs from the healthy twin against the patient’s autologous bone marrow (BM) blasts, mediated by SPM-2. The NCR receptor expression profiles on NKs from patient and twin were similar, but NK cell titers in peripheral blood were lower for samples drawn at diagnosis than in remission. Conclusions Peripheral blood NK titers and ex vivo cytolytic activities mediated by triplebody SPM-2 were comparable for cells drawn from an AML patient in remission and a healthy twin. If these results can be generalized, then NKs from AML patients in remission are sufficient in numbers and cytolytic activity to make triplebodies promising new agents for the treatment of AML.
Collapse
Affiliation(s)
- Todd A Braciak
- Division of Hematology and Oncology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ziemssenstrasse 1, D-80336 Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Preferential eradication of acute myelogenous leukemia stem cells by fenretinide. Proc Natl Acad Sci U S A 2013; 110:5606-11. [PMID: 23513221 DOI: 10.1073/pnas.1302352110] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Leukemia stem cells (LSCs) play important roles in leukemia initiation, progression, and relapse, and thus represent a critical target for therapeutic intervention. However, relatively few agents have been shown to target LSCs, slowing progress in the treatment of acute myelogenous leukemia (AML). Based on in vitro and in vivo evidence, we report here that fenretinide, a well-tolerated vitamin A derivative, is capable of eradicating LSCs but not normal hematopoietic progenitor/stem cells at physiologically achievable concentrations. Fenretinide exerted a selective cytotoxic effect on primary AML CD34(+) cells, especially the LSC-enriched CD34(+)CD38(-) subpopulation, whereas no significant effect was observed on normal counterparts. Methylcellulose colony formation assays further showed that fenretinide significantly suppressed the formation of colonies derived from AML CD34(+) cells but not those from normal CD34(+) cells. Moreover, fenretinide significantly reduced the in vivo engraftment of AML stem cells but not normal hematopoietic stem cells in a nonobese diabetic/SCID mouse xenotransplantation model. Mechanistic studies revealed that fenretinide-induced cell death was linked to a series of characteristic events, including the rapid generation of reactive oxygen species, induction of genes associated with stress responses and apoptosis, and repression of genes involved in NF-κB and Wnt signaling. Further bioinformatic analysis revealed that the fenretinide-down-regulated genes were significantly correlated with the existing poor-prognosis signatures in AML patients. Based on these findings, we propose that fenretinide is a potent agent that selectively targets LSCs, and may be of value in the treatment of AML.
Collapse
|
18
|
Ma Y, Conforti R, Aymeric L, Locher C, Kepp O, Kroemer G, Zitvogel L. How to improve the immunogenicity of chemotherapy and radiotherapy. Cancer Metastasis Rev 2011; 30:71-82. [PMID: 21298323 DOI: 10.1007/s10555-011-9283-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chemotherapy or radiotherapy could induce various tumor cell death modalities, releasing tumor-derived antigen as well as danger signals that could either be captured for triggering antitumor immune response or ignored. Exploring the interplay among therapeutic drugs, tumor cell death and the immune cells should improve diagnostic, prognostic, predictive, and therapeutic management of tumor. We summarized some of the cell death-derived danger signals and the mechanism for host to sense and response to cell death in the tumor microenvironment. Based on the recent clinical or experimental findings, several strategies have been suggested to improve the immunogenicity of cell death and augment antitumor immunity.
Collapse
Affiliation(s)
- Yuting Ma
- INSERM, U1015, 94805 Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
19
|
Ishikawa F. ["Creation of mouse models for human diseases"]. NIHON RINSHO MEN'EKI GAKKAI KAISHI = JAPANESE JOURNAL OF CLINICAL IMMUNOLOGY 2011; 33:304-11. [PMID: 21212582 DOI: 10.2177/jsci.33.304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
To translate research findings into medicine and drug development, we have been performing in vivo research using primary human samples. Development of mouse models with reconstituted human immunity would be critical to overcome technical and ethical constraints associated with in vivo human research. To this end, we have created humanized mice by intravenously injecting purified human hematopoietic stem cells (HSCs) into immune-compromised NOD/SCID/IL2rgKO newborns. This xenogeneic transplantation system allows long-term engraftment and multi-lineage differentiation of human HSCs. The humanized mouse fully reconstituted with human myeloid and lymphoid subsets is expected to serve as an in vivo platform for the investigation of human immune function. In addition to understanding normal human hematopoiesis and immunity, the use of humanized mice is expected to allow investigators to translate their research findings into therapeutic and pharmaceutical development. As a possibility for such translation, we developed an in vivo model of human acute myeloid leukemia (AML), a disease in which the majority of patients succumb to relapse. Using the model, we examined the pathogenesis of AML and tried to clarify the role of AML stem cells in chemotherapy resistance and relapse. Humanized mouse model for both normal and diseased immuno-hematopoietic system is opening a new era in translational medicine.
Collapse
Affiliation(s)
- Fumihiko Ishikawa
- RIKEN Research Center for Allergy & Immunology Research Unit for Human Disease Models
| |
Collapse
|
20
|
Abstract
Recently, several research groups have described cell surface molecules that are selectively or differentially expressed on leukemia stem cells (LSCs) relative to normal tissue. The identification of these antigens suggests that antibody-based diagnostic or therapeutic opportunities may be forthcoming. Indeed, preclinical studies have suggested the utility of targeting such molecules as a means of enhancing leukemia therapy. Here the current understanding of the LSC phenotype is described, and the potential application of antibody-based treatment regimens is discussed.
Collapse
Affiliation(s)
- Craig T Jordan
- Departments of Medicine and Biomedical Genetics and the James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|