1
|
Wang Y, Xiu Y, Dong Q, Zhao J, Neumbo K, Miyagi M, Borcherding N, Fu L, De Celis H, Pintozzi N, Starczynowski DT, Zhao C. TIFAB modulates metabolic pathways in KMT2A::MLLT3-induced AML through HNF4A. Blood Adv 2025; 9:844-855. [PMID: 39626355 PMCID: PMC11872587 DOI: 10.1182/bloodadvances.2024013446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/11/2024] [Indexed: 03/05/2025] Open
Abstract
ABSTRACT Tumor necrosis factor (TNF) receptor-associated factor (TRAF)-interacting protein with forkhead-associated domain B (TIFAB), an inhibitor of NF-κB signaling, plays critical roles in hematopoiesis, myelodysplastic neoplasms, and leukemia. We previously demonstrated that Tifab enhances KMT2A::MLLT3-driven acute myeloid leukemia (AML) by either upregulating Hoxa9 or through ubiquitin-specific peptidase 15-mediated downregulation of p53 signaling. In this study, we show that Tifab deletion in KMT2A::MLLT3-induced AML impairs leukemia stem/progenitor cell (LSPC) engraftment, glucose uptake, and mitochondrial function. Gene set enrichment analysis reveals that Tifab deletion downregulates MYC, HOXA9/MEIS1, mTORC1 signaling, and genes involved in glycolysis and oxidative phosphorylation. By comparing genes upregulated in TIFAB-overexpressing LSPCs with those downregulated upon Tifab deletion, we identify hepatocyte nuclear factor 4 alpha (Hnf4a) as a key TIFAB target, regulated through the inhibition of NF-κB component RelB, which suppresses Hnf4a in leukemia cells. HNF4A, a nuclear receptor involved in organ development, metabolism, and tumorigenesis, rescues the metabolic defects caused by Tifab deletion and enhances leukemia cell engraftment. Conversely, Hnf4a knockdown attenuates TIFAB-mediated enhancement of LSPC function. These findings highlight the critical role of the TIFAB-HNF4A axis in KMT2A::MLLT3-induced AML and uncover a novel regulator in leukemia biology.
Collapse
Affiliation(s)
- Yang Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Yan Xiu
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Qianze Dong
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | - Jinming Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, OH
- Department of Pathology, China Medical University, Shenyang, China
| | - Kelao Neumbo
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH
| | - Masaru Miyagi
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Lin Fu
- Department of Pathology, China Medical University, Shenyang, China
| | - Havana De Celis
- Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Nicolas Pintozzi
- Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Daniel T. Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- University of Cincinnati Cancer Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH
| | - Chen Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, OH
- Department of Pathology, University Hospitals Case Medical Center, Cleveland, OH
- Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH
| |
Collapse
|
2
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Jiang X. RNA modification in normal hematopoiesis and hematologic malignancies. MedComm (Beijing) 2024; 5:e787. [PMID: 39445003 PMCID: PMC11496571 DOI: 10.1002/mco2.787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
N6-methyladenosine (m6A) is the most abundant RNA modification in eukaryotic cells. Previous studies have shown that m6A plays a critical role under both normal physiological and pathological conditions. Hematopoiesis and differentiation are highly regulated processes, and recent studies on m6A mRNA methylation have revealed how this modification controls cell fate in both normal and malignant hematopoietic states. However, despite these insights, a comprehensive understanding of its complex roles between normal hematopoietic development and malignant hematopoietic diseases remains elusive. This review first provides an overview of the components and biological functions of m6A modification regulators. Additionally, it highlights the origin, differentiation process, biological characteristics, and regulatory mechanisms of hematopoietic stem cells, as well as the features, immune properties, and self-renewal pathways of leukemia stem cells. Last, the article systematically reviews the latest research advancements on the roles and mechanisms of m6A regulatory factors in normal hematopoiesis and related malignant diseases. More importantly, this review explores how targeting m6A regulators and various signaling pathways could effectively intervene in the development of leukemia, providing new insights and potential therapeutic targets. Targeting m6A modification may hold promise for achieving more precise and effective leukemia treatments.
Collapse
Affiliation(s)
- Xi Chen
- Department of NeurosurgeryThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingYunnanChina
| | - Yixiao Yuan
- Department of MedicineUF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
- Department of Medicine and Department of Biochemistry and Molecular BiologyUniversity of FloridaGainesvilleFloridaUSA
| | - Fan Zhou
- Department of NeurosurgeryThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingYunnanChina
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingYunnanChina
| | - Jun Pu
- Department of NeurosurgeryThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingYunnanChina
| | - Xiulin Jiang
- Department of MedicineUF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
- Department of Medicine and Department of Biochemistry and Molecular BiologyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
3
|
Lomov NA, Viushkov VS, Rubtsov MA. Mechanisms of Secondary Leukemia Development Caused by Treatment with DNA Topoisomerase Inhibitors. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:892-911. [PMID: 37751862 DOI: 10.1134/s0006297923070040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 09/28/2023]
Abstract
Leukemia is a blood cancer originating in the blood and bone marrow. Therapy-related leukemia is associated with prior chemotherapy. Although cancer therapy with DNA topoisomerase II inhibitors is one of the most effective cancer treatments, its side effects include development of secondary leukemia characterized by the chromosomal rearrangements affecting AML1 or MLL genes. Recurrent chromosomal translocations in the therapy-related leukemia differ from chromosomal rearrangements associated with other neoplasias. Here, we reviewed the factors that drive chromosomal translocations induced by cancer treatment with DNA topoisomerase II inhibitors, such as mobility of ends of double-strand DNA breaks formed before the translocation and gain of function of fusion proteins generated as a result of translocation.
Collapse
Affiliation(s)
- Nikolai A Lomov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Vladimir S Viushkov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Mikhail A Rubtsov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Department of Biochemistry, Center for Industrial Technologies and Entrepreneurship Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| |
Collapse
|
4
|
Zhang J, Oak J. Challenges of detecting measurable/minimal disease in acute leukemia. Semin Diagn Pathol 2023; 40:216-220. [PMID: 37150656 DOI: 10.1053/j.semdp.2023.04.004] [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] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023]
Abstract
Measurable/minimal residual disease (MRD) tracking has emerged as a powerful tool for assessing treatment response and predicting outcomes in acute leukemia. However, the clinical and technological challenges associated with MRD tracking must be addressed to improve its utility in routine patient care. This review article aims to provide a summary of the different MRD methodologies used in acute leukemia. It highlights the strengths, diagnostic pitfalls, and clinical utility associated with MRD tracking in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Pathology, 300 Pasteur Drive, L235, Stanford, CA 94305, United States
| | - Jean Oak
- Department of Pathology, 300 Pasteur Drive, L235, Stanford, CA 94305, United States.
| |
Collapse
|
5
|
Dong Q, Xiu Y, Wang Y, Hodgson C, Borcherding N, Jordan C, Buchanan J, Taylor E, Wagner B, Leidinger M, Holman C, Thiele DJ, O’Brien S, Xue HH, Zhao J, Li Q, Meyerson H, Boyce BF, Zhao C. HSF1 is a driver of leukemia stem cell self-renewal in acute myeloid leukemia. Nat Commun 2022; 13:6107. [PMID: 36245043 PMCID: PMC9573868 DOI: 10.1038/s41467-022-33861-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 10/05/2022] [Indexed: 01/25/2023] Open
Abstract
Acute myeloid leukemia (AML) is maintained by self-renewing leukemic stem cells (LSCs). A fundamental problem in treating AML is that conventional therapy fails to eliminate LSCs, which can reinitiate leukemia. Heat shock transcription factor 1 (HSF1), a central regulator of the stress response, has emerged as an important target in cancer therapy. Using genetic Hsf1 deletion and a direct HSF1 small molecule inhibitor, we show that HSF1 is specifically required for the maintenance of AML, while sparing steady-state and stressed hematopoiesis. Mechanistically, deletion of Hsf1 dysregulates multifaceted genes involved in LSC stemness and suppresses mitochondrial oxidative phosphorylation through downregulation of succinate dehydrogenase C (SDHC), a direct HSF1 target. Forced expression of SDHC largely restores the Hsf1 ablation-induced AML developmental defect. Importantly, the growth and engraftment of human AML cells are suppressed by HSF1 inhibition. Our data provide a rationale for developing efficacious small molecules to specifically target HSF1 in AML.
Collapse
Affiliation(s)
- Qianze Dong
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA
| | - Yan Xiu
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA ,grid.410349.b0000 0004 5912 6484Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106 USA
| | - Yang Wang
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA
| | | | - Nick Borcherding
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Craig Jordan
- grid.241116.10000000107903411Division of Hematology, University of Colorado Anschutz Campus, Denver, CO 80045 USA
| | - Jane Buchanan
- grid.214572.70000 0004 1936 8294Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52240 USA
| | - Eric Taylor
- grid.214572.70000 0004 1936 8294Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52240 USA
| | - Brett Wagner
- grid.214572.70000 0004 1936 8294Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242 USA
| | - Mariah Leidinger
- grid.214572.70000 0004 1936 8294Department of Pathology, University of Iowa, Iowa City, IA 52242 USA
| | - Carol Holman
- grid.214572.70000 0004 1936 8294Department of Pathology, University of Iowa, Iowa City, IA 52242 USA
| | | | | | - Hai-hui Xue
- grid.239835.60000 0004 0407 6328Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110 USA
| | - Jinming Zhao
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA ,grid.412449.e0000 0000 9678 1884Department of Pathology, China Medical University, 77 Puhe Rd, Shenbei Xinqu, Shenyang Shi, 110122 Liaoning Sheng China
| | - Qingchang Li
- grid.412449.e0000 0000 9678 1884Department of Pathology, China Medical University, 77 Puhe Rd, Shenbei Xinqu, Shenyang Shi, 110122 Liaoning Sheng China
| | - Howard Meyerson
- grid.443867.a0000 0000 9149 4843Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 USA
| | - Brendan F. Boyce
- grid.412750.50000 0004 1936 9166Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Chen Zhao
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA ,grid.410349.b0000 0004 5912 6484Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106 USA ,grid.443867.a0000 0000 9149 4843Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 USA
| |
Collapse
|
6
|
Shrestha S, Banstola A, Jeong JH, Seo JH, Yook S. Targeting Cancer Stem Cells: Therapeutic and diagnostic strategies by the virtue of nanoparticles. J Control Release 2022; 348:518-536. [PMID: 35709876 DOI: 10.1016/j.jconrel.2022.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 12/18/2022]
Abstract
Cancer stem cells (CSCs) are the subpopulation of cells present within a tumor with the properties of self-renewing, differentiating, and proliferating. Owing to the presence of ATP-binding cassette drug pumps and increased expression of anti-apoptotic proteins, the conventional chemotherapeutic agents have failed to eliminate CSCs resulting in relapse and resistance of cancer. Therefore, to obtain long-lasting clinical responses and avoid the recurrence of cancer, it is crucial to develop an efficient strategy targeting CSCs by either employing a differentiation therapy or specifically delivering drugs to CSCs. Several intracellular and extracellular cancer specific biomarkers are overexpressed by CSCs and are utilized as targets for the development of new approaches in the diagnosis and treatment of CSCs. Moreover, several nanostructured particles, alone or in combination with current treatment approaches, have been used to improve the detection, imaging, and targeting of CSCs, thus addressing the limitations of cancer therapies. Targeting CSC surface markers, stemness-related signaling pathways, and tumor microenvironmental signals has improved the detection and eradication of CSCs and, therefore, tumor diagnosis and treatment. This review summarizes a variety of promising nanoparticles targeting the surface biomarkers of CSCs for the detection and eradication of tumor-initiating stem cells, used in combination with other treatment regimens.
Collapse
Affiliation(s)
- Samjhana Shrestha
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea
| | - Asmita Banstola
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea; Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Hae Seo
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea.
| |
Collapse
|
7
|
Ngai LL, Kelder A, Janssen JJWM, Ossenkoppele GJ, Cloos J. MRD Tailored Therapy in AML: What We Have Learned So Far. Front Oncol 2021; 10:603636. [PMID: 33575214 PMCID: PMC7871983 DOI: 10.3389/fonc.2020.603636] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/16/2020] [Indexed: 12/22/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal disease associated with a dismal survival, partly due to the frequent occurrence of relapse. Many patient- and leukemia-specific characteristics, such as age, cytogenetics, mutations, and measurable residual disease (MRD) after intensive chemotherapy, have shown to be valuable prognostic factors. MRD has become a rich field of research where many advances have been made regarding technical, biological, and clinical aspects, which will be the topic of this review. Since many laboratories involved in AML diagnostics have experience in immunophenotyping, multiparameter flow cytometry (MFC) based MRD is currently the most commonly used method. Although molecular, quantitative PCR based techniques may be more sensitive, their disadvantage is that they can only be applied in a subset of patients harboring the genetic aberration. Next-generation sequencing can assess and quantify mutations in many genes but currently does not offer highly sensitive MRD measurements on a routine basis. In order to provide reliable MRD results, MRD assay optimization and standardization is essential. Different techniques for MRD assessment are being evaluated, and combinations of the methods have shown promising results for improving its prognostic value. In this regard, the load of leukemic stem cells (LSC) has also been shown to add to the prognostic value of MFC-MRD. At this moment, MRD after intensive chemotherapy is most often used as a prognostic factor to help stratify patients, but also to select the most appropriate consolidation therapy. For example, to guide post-remission treatment for intermediate-risk patients where MRD positive patients receive allogeneic stem cell transplantation and MRD negative receive autologous stem cell transplantation. Other upcoming uses of MRD that are being investigated include: selecting the type of allogeneic stem cell transplantation therapy (donor, conditioning), monitoring after stem cell transplantation (to allow intervention), and determining drug efficacy for the use of a surrogate endpoint in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Jacqueline Cloos
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit, Amsterdam, Netherlands
| |
Collapse
|
8
|
Zeijlemaker W, Kelder A, Cloos J, Schuurhuis GJ. Immunophenotypic Detection of Measurable Residual (Stem Cell) Disease Using LAIP Approach in Acute Myeloid Leukemia. CURRENT PROTOCOLS IN CYTOMETRY 2019; 91:e66. [PMID: 31763792 PMCID: PMC6856793 DOI: 10.1002/cpcy.66] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Half of the patients with acute myeloid leukemia (AML), who achieve complete remission after chemotherapy treatment, will ultimately experience a relapse. Measurable residual disease (MRD) is an important post-treatment risk factor in AML, because it gives additional information about the depth of the remission. Within MRD, the small population of leukemic stem cells (LSCs) is thought to be at the base of the actual relapse. In this protocol, the flow cytometric detection of MRD and LSCs herein is outlined. We give a detailed overview of the sampling procedures for optimal multiparameter flow cytometry assessment of both MRD and LSC, using leukemia associated immunophenotypes (LAIPs) and LSC markers. Moreover, an overview of the gating strategies to detect LAIPs and LSC markers is provided. This protocol serves as guidance for flow cytometric detection of measurable residual (stem cell) disease necessary for proper therapeutic decision making in AML patients. © 2019 The Authors. Basic Protocol 1: Immunophenotypic LAIP detection for measurable residual disease monitoring Basic Protocol 2: Immunophenotypic detection of CD34+CD38- leukemic stem cells.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/metabolism
- Antigens, CD34/metabolism
- Biomarkers, Tumor/analysis
- Bone Marrow Cells/pathology
- Cell Count
- Cells, Cultured
- Flow Cytometry/methods
- Humans
- Immunophenotyping/methods
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Monitoring, Physiologic/methods
- Neoplasm, Residual
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Recurrence
Collapse
Affiliation(s)
- Wendelien Zeijlemaker
- Department of HematologyAmsterdam University Medical Center, Cancer Center VU University Medical CenterAmsterdamThe Netherlands
| | - Angele Kelder
- Department of HematologyAmsterdam University Medical Center, Cancer Center VU University Medical CenterAmsterdamThe Netherlands
| | - Jacqueline Cloos
- Department of HematologyAmsterdam University Medical Center, Cancer Center VU University Medical CenterAmsterdamThe Netherlands
| | - Gerrit Jan Schuurhuis
- Department of HematologyAmsterdam University Medical Center, Cancer Center VU University Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
9
|
Development of an anti-CD45RA-quantum dots conjugated scFv to detect leukemic cancer stem cells. Mol Biol Rep 2019; 47:225-234. [DOI: 10.1007/s11033-019-05122-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 10/09/2019] [Indexed: 11/26/2022]
|
10
|
Leukemia Stem Cells in the Pathogenesis, Progression, and Treatment of Acute Myeloid Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:95-128. [DOI: 10.1007/978-981-13-7342-8_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
11
|
Zeijlemaker W, Grob T, Meijer R, Hanekamp D, Kelder A, Carbaat-Ham JC, Oussoren-Brockhoff YJM, Snel AN, Veldhuizen D, Scholten WJ, Maertens J, Breems DA, Pabst T, Manz MG, van der Velden VHJ, Slomp J, Preijers F, Cloos J, van de Loosdrecht AA, Löwenberg B, Valk PJM, Jongen-Lavrencic M, Ossenkoppele GJ, Schuurhuis GJ. CD34 +CD38 - leukemic stem cell frequency to predict outcome in acute myeloid leukemia. Leukemia 2018; 33:1102-1112. [PMID: 30542144 DOI: 10.1038/s41375-018-0326-3] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 12/29/2022]
Abstract
Current risk algorithms are primarily based on pre-treatment factors and imperfectly predict outcome in acute myeloid leukemia (AML). We introduce and validate a post-treatment approach of leukemic stem cell (LSC) assessment for prediction of outcome. LSC containing CD34+CD38- fractions were measured using flow cytometry in an add-on study of the HOVON102/SAKK trial. Predefined cut-off levels were prospectively evaluated to assess CD34+CD38-LSC levels at diagnosis (n = 594), and, to identify LSClow/LSChigh (n = 302) and MRDlow/MRDhigh patients (n = 305) in bone marrow in morphological complete remission (CR). In 242 CR patients combined MRD and LSC results were available. At diagnosis the CD34+CD38- LSC frequency independently predicts overall survival (OS). After achieving CR, combining LSC and MRD showed reduced survival in MRDhigh/LSChigh patients (hazard ratio [HR] 3.62 for OS and 5.89 for cumulative incidence of relapse [CIR]) compared to MRDlow/LSChigh, MRDhigh/LSClow, and especially MRDlow/LSClow patients. Moreover, in the NPM1mutant positive sub-group, prognostic value of golden standard NPM1-MRD by qPCR can be improved by addition of flow cytometric approaches. This is the first prospective study demonstrating that LSC strongly improves prognostic impact of MRD detection, identifying a patient subgroup with an almost 100% treatment failure probability, warranting consideration of LSC measurement incorporation in future AML risk schemes.
Collapse
Affiliation(s)
- Wendelien Zeijlemaker
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Tim Grob
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rosa Meijer
- Clinical trial Center- HOVON data center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Diana Hanekamp
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Angèle Kelder
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jannemieke C Carbaat-Ham
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Alexander N Snel
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Dennis Veldhuizen
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Willemijn J Scholten
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Johan Maertens
- Department of Hematology, University Hospitals Leuven, Campus Gasthuisberg, Leuven, Belgium
| | - Dimitri A Breems
- Department of Hematology, Ziekenhuis Netwerk Antwerpen, Antwerp, Belgium
| | - Thomas Pabst
- Department of Hematology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Markus G Manz
- Department of Hematology, University and University Hospital Zürich, Zürich, Switzerland
| | | | - Jennichjen Slomp
- Department of Clinical Chemistry, Medisch Spectrum Twente/Medlon, Enschede, The Netherlands
| | - Frank Preijers
- Department of Laboratory Medicine - Laboratory for Hematology, Radboud University Nijmegen Medical Center, RUNMC, Nijmegen, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Oncology/Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Arjan A van de Loosdrecht
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Bob Löwenberg
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter J M Valk
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gerrit J Schuurhuis
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
12
|
Studencka-Turski M, Maubach G, Feige MH, Naumann M. Constitutive activation of nuclear factor kappa B-inducing kinase counteracts apoptosis in cells with rearranged mixed lineage leukemia gene. Leukemia 2018; 32:2498-2501. [PMID: 29725033 DOI: 10.1038/s41375-018-0128-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/14/2018] [Accepted: 03/29/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Maja Studencka-Turski
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Gunter Maubach
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Michael Hartmut Feige
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Magdeburg, Germany.
| |
Collapse
|
13
|
Cornet-Masana JM, Moreno-Martínez D, Lara-Castillo MC, Nomdedeu M, Etxabe A, Tesi N, Pratcorona M, Esteve J, Risueño RM. Emetine induces chemosensitivity and reduces clonogenicity of acute myeloid leukemia cells. Oncotarget 2018; 7:23239-50. [PMID: 26992240 PMCID: PMC5029623 DOI: 10.18632/oncotarget.8096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 02/28/2016] [Indexed: 01/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is an hematologic neoplasia characterized by the accumulation of transformed immature myeloid cells in bone marrow. Although the response rate to induction therapy is high, survival rate 5-year after diagnosis is still low, highlighting the necessity of new novel agents. To identify agents with the capability to abolish the self-renewal capacity of AML blasts, an in silico screening was performed to search for small molecules that induce terminal differentiation. Emetine, a hit compound, was validated for its anti-leukemic effect in vitro, ex vivo and in vivo. Emetine, a second-line anti-protozoa drug, differentially reduced cell viability and clonogenic capacity of AML primary patient samples, sparing healthy blood cells. Emetine treatment markedly reduced AML burden in bone marrow of xenotransplanted mice and decreased self-renewal capacity of the remaining engrafted AML cells. Emetine also synergized with commonly used chemotherapeutic agents such as ara-C. At a molecular level, emetine treatment was followed by a reduction in HIF-1α protein levels. This study validated the anti-leukemiceffect of emetine in AML cell lines, a group of diverse AML primary samples, and in a human AML-transplanted murine model, sparing healthy blood cells. The selective anti-leukemic effect of emetine together with the safety of the dose range required to exert this effect support the development of this agent in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Meritxell Nomdedeu
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Amaia Etxabe
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Niccolò Tesi
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Marta Pratcorona
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Esteve
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ruth M Risueño
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| |
Collapse
|
14
|
Liu FR, Jin H, Wang Y, Chen C, Li M, Mao SJ, Wang Q, Li H. Anti-CD123 antibody-modified niosomes for targeted delivery of daunorubicin against acute myeloid leukemia. Drug Deliv 2017; 24:882-890. [PMID: 28574300 PMCID: PMC8244627 DOI: 10.1080/10717544.2017.1333170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
A novel niosomal delivery system was designed and investigated for the targeted delivery of daunorubicin (DNR) against acute myeloid leukemia (AML). Anti-CD123 antibodies conjugated to Mal-PEG2000-DSPE were incorporated into normal niosomes (NS) via a post insertion method to afford antibody-modified niosomes (CD123-NS). Next, NS was modified with varying densities of antibody (0.5 or 2%, antibody/Span 80, molar ratio), thus providing L-CD123-NS and H-CD123-NS. We studied the effect of antibody density on the uptake efficiency of niosomes in NB4 and THP-1 cells, on which CD123 express differently. Our results demonstrate CD123-NS showed significantly higher uptake efficiency than NS in AML cells, and the uptake efficiency of CD123-NS has been ligand density-dependent. Also, AML cells preincubated with anti-CD123 antibody showed significantly reduced cellular uptake of CD123-NS compared to control. Further study on the uptake mechanism confirmed a receptor-mediated endocytic process. Daunorubicin (DNR)-loaded H-CD123-NS demonstrated a 2.45- and 3.22-fold higher cytotoxicity, compared to DNR-loaded NS in NB4 and THP-1 cells, respectively. Prolonged survival time were observed in leukemic mice treated with DNR-H-CD123-NS. Collectively, these findings support that the CD123-NS represent a promising delivery system for the treatment of AML.
Collapse
Affiliation(s)
- Fu-Rong Liu
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education and West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Hui Jin
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education and West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Yin Wang
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education and West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Chen Chen
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education and West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Ming Li
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education and West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Sheng-Jun Mao
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education and West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Qiantao Wang
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education and West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Hui Li
- b Department of Hematology , Sichuan Academy of Medical Sciences and Sichuan Provincial People Hospital , Chengdu , China
| |
Collapse
|
15
|
Mukherjee D, Kundu N, Chakravarty L, Behera B, Chakrabarti P, Sarkar N, Maiti TK. Membrane perturbation through novel cell-penetrating peptides influences intracellular accumulation of imatinib mesylate in CML cells. Cell Biol Toxicol 2017; 34:233-245. [PMID: 29046997 DOI: 10.1007/s10565-017-9414-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/18/2017] [Indexed: 12/17/2022]
Abstract
Chronic myeloid leukemia is a stem cell disease with the presence of Philadelphia chromosome generated through reciprocal translocation of chromosome 9 and 22. The use of first- and second-generation tyrosine kinase inhibitors has been successful to an extent. However, resistance against such drugs is an emerging problem. Apart from several drug-resistant mechanisms, drug influx/efflux ratio appears to be one of the key determinants of therapeutic outcomes. In addition, intracellular accumulation of drug critically depends on cell membrane fluidity and lipid raft dynamics. Previously, we reported two novel cell-penetrating peptides (CPPs), namely, cationic IR15 and anionic SR11 present in tryptic digest of Abrus agglutinin. Here, the potential of IR15 and SR11 to influence intracellular concentration of imatinib has been evaluated. Fluorescent correlation spectroscopy and lifetime imaging were employed to map membrane fluidity and lipid raft distribution following peptide-drug co-administration. Results show that IR15 and SR11 are the two CPPs which can modulate membrane fluidity and lipid raft distribution in K562 cells. Both IR15 and SR11 significantly reduce the viability of CML cells in the presence of imatinib by increasing the intracellular accumulation of the drug.
Collapse
Affiliation(s)
- Devdeep Mukherjee
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Niloy Kundu
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Lopamudra Chakravarty
- Department of Haematology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Birendra Behera
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Prantar Chakrabarti
- Department of Haematology, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - Nilmoni Sarkar
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| |
Collapse
|
16
|
Madhumathi J, Sridevi S, Verma RS. CD25 targeted therapy of chemotherapy resistant leukemic stem cells using DR5 specific TRAIL peptide. Stem Cell Res 2017; 19:65-75. [DOI: 10.1016/j.scr.2017.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/24/2016] [Accepted: 01/03/2017] [Indexed: 12/13/2022] Open
|
17
|
Espirito Santo A, Chacim S, Ferreira I, Leite L, Moreira C, Pereira D, Dantas M, Nunes M, Viterbo L, Moreira I, Martins A, Oliveira I, Domingues N, Mariz J, Medeiros R. Southwestern Oncology Group pretreatment risk criteria as predictive or prognostic factors in acute myeloid leukemia. Mol Clin Oncol 2017; 6:384-388. [PMID: 28451418 DOI: 10.3892/mco.2017.1134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 07/25/2017] [Indexed: 11/05/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal hematological malignant condition and the implications of pretreatment risk criteria as predictive or prognostic factors are constantly under evaluation. With this study, the authors' intent was to characterize AML patients and to evaluate the clinical outcome associated with Southwestern Oncology Group (SWOG) coding pretreatment risk criteria/cytogenetic score. Between 2002 and 2010, 225 patients were diagnosed with AML at the Portuguese Institute of Oncology (Porto, Portugal). From this patient group, 128 patients aged <65 years were selected. The patients were treated using a combination of cytarabine and anthracycline, with the addition of cyclosporine when bone marrow dysplasia was observed. A median survival of 24 months was observed in this group. The patients were divided in subgroups according to the SWOG pretreatment risk criteria. We observed a statistically significant association of non-favorable SWOG coding with female gender [P=0.025; risk ratio (RR)=3.632, 95% confidence interval (CI): 1.113-11.852], indication for allogeneic bone marrow transplantation (P=0.023, RR=1.317, 95% CI: 1.184-1.465), complete response achievement (P=0.013, RR=1.385, 95% CI: 11.232-1.556) and relapse (P=0.048, RR=3.181, 95% CI: 10.966-10.478). Furthermore, SWOG pretreatment risk criteria also significantly affected global overall survival (OS; P=0.003) and OS at 5 years (P=0.001). A multivariate Cox regression analysis supported response to induction therapy (3-year OS: P=0.011, RR=0.385, 95% CI: 10.184-0.806; 5-year OS: P=0.012, RR=0.388, 95% CI: 10.597-1.994), consolidation (3-year OS: P=0.005, RR=0.328, 95% CI: 0.150-0.720; 5-year OS: P=0.002, RR=0.308, 95% CI: 0.144-0.657) and the diagnosis of therapy-related aml (3-year OS: P=0.016, RR=2.756, 95% CI: 0.486-1.281; 5-year OS: P=0.031, RR=2.369, 95% CI: 1.081-5.189) as prognostic factors, but this was not confirmed for SWOG pretreatment risk criteria. Therefore, we concluded that the reproducibility of the application of the SWOG pretreatment risk criteria may not be available as a prognostic factor in every acute leukemia population. However, its application as a predictive factor of response has been confirmed in our population.
Collapse
Affiliation(s)
- Ana Espirito Santo
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal.,Faculty of Medicine, University of Porto, 4000-286 Porto, Portugal
| | - Sergio Chacim
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Isabel Ferreira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Luis Leite
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Claudia Moreira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Dulcineia Pereira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Margarida Dantas
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Marta Nunes
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Luisa Viterbo
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Ilidia Moreira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Angelo Martins
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Isabel Oliveira
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Nelson Domingues
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Jose Mariz
- Department of Onco-Hematology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology Group-CI, Portuguese Institute of Oncology, 4200-072 Porto, Portugal.,Abel Salazar Institute of Biomedical Sciences, University of Porto, 4000-286 Porto, Portugal.,Oncology Department, Portuguese Institute of Oncology, 4200-072 Porto, Portugal.,Biomedical Research Center, Faculty of Health Sciences, Fernando Pessoa University, 4000-286 Porto, Portugal.,Research Department, Portuguese League Against Cancer, 4200-072 Porto, Portugal
| |
Collapse
|
18
|
Hu J, Feng M, Liu ZL, Liu Y, Huang ZL, Li H, Feng WL. Potential role of Wnt/β-catenin signaling in blastic transformation of chronic myeloid leukemia: cross talk between β-catenin and BCR-ABL. Tumour Biol 2016; 37:15859–15872. [PMID: 27817074 DOI: 10.1007/s13277-016-5413-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/13/2016] [Indexed: 12/25/2022] Open
Abstract
Chronic myeloid leukemia (CML) results from malignant transformation of hematopoietic stem cells induced by the BCR-ABL oncogene. Transformation from chronic to blastic phase is the lethal step in CML. Leukemic stem cells (LSCs) are the basic reason for blastic transformation. It has been shown that Wnt/β-catenin signaling contributes to the self-renewal capacity and proliferation of LSCs in CML. However, the role of Wnt/β-catenin signaling in blastic transformation of CML is still obscure. Here, we explored the relationship between BCR-ABL and β-catenin signaling in vitro and in vivo. We found that BCR-ABL stimulated β-catenin via activation of PI3K/AKT signaling in blastic phase CML cells. Inhibition of the kinase activity of BCR-ABL, PI3K, or AKT decreased the level of β-catenin in both K562 cells and a CML mouse model and suppressed the transcription of downstream target genes (c-myc and cyclin D1). In addition, inhibition of the BCR-ABL/PI3K/AKT pathway delayed the disease progression in the CML mouse model. To further explore the role of β-catenin in the self-renewal and survival of CML LSCs, we established a secondary transplantation CML mouse model. Our data revealed that inhibition of the BCR-ABL/PI3K/AKT pathway reduced the tumor-initiating ability of K562 cells, decreased leukemia cell infiltration into peripheral blood and bone marrow, and prolonged the survival of mice. In conclusion, our data indicate a close relationship between β-catenin and BCR-ABL/PI3K/AKT in blastic phase CML. β-Catenin inhibition may be of therapeutic value by targeting LSCs in combination with a tyrosine kinase inhibitor, which may delay blastic transformation of CML.
Collapse
Affiliation(s)
- Jing Hu
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Min Feng
- Institute of Neuroscience, Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, China
| | - Zhang-Ling Liu
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Yi Liu
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Zheng-Lan Huang
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Hui Li
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Wen-Li Feng
- Department of Clinical Hematology, Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
19
|
Li B, Zhao W, Zhang X, Wang J, Luo X, Baker SD, Jordan CT, Dong Y. Design, synthesis and evaluation of anti-CD123 antibody drug conjugates. Bioorg Med Chem 2016; 24:5855-5860. [PMID: 27687970 DOI: 10.1016/j.bmc.2016.09.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 01/02/2023]
Abstract
Leukemia stem cells (LSCs) account for the development of drug resistance and increased recurrence rate in acute myeloid leukemia (AML) patients. Targeted drug delivery to leukemia stem cells remains a major challenge in AML chemotherapy. Overexpressed interleukin-3 receptor alpha chain, CD123, on the surface of leukemia stem cells was reported to be a potential target in AML treatment. Here, we designed and developed an antibody drug conjugate (CD123-CPT) by integrating anti-CD123 antibody with a chemotherapeutic agent, Camptothecin (CPT), via a disulfide linker. The linker is biodegradable in the presence of Glutathione (GSH, an endogenous component in cells), which leads to release of CPT. Anti-CD123 antibody conjugates showed significant higher cellular uptake in CD123-overexpressed tumor cells. More importantly, CD123-CPT demonstrated potent inhibitory effects on CD123-overexpressed tumor cells. Consequently, these results provide a promising targeted chemotherapeutical strategy for AML treatment.
Collapse
Affiliation(s)
- Bin Li
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Weiyu Zhao
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Xinfu Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Junfeng Wang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Xiao Luo
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Sharyn D Baker
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Craig T Jordan
- Division of Hematology, University of Colorado, Aurora, CO 80045, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
20
|
Zeng Y, Weng G, Fan J, Li Z, Wu J, Li Y, Zheng R, Xia P, Guo K. Curcumin reduces the expression of survivin, leading to enhancement of arsenic trioxide-induced apoptosis in myelodysplastic syndrome and leukemia stem-like cells. Oncol Rep 2016; 36:1233-42. [PMID: 27430728 PMCID: PMC5001835 DOI: 10.3892/or.2016.4944] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/19/2016] [Indexed: 12/29/2022] Open
Abstract
Low response, treatment-related complications and relapse due to the low sensitivity of myelodysplastic syndrome (MDS) and leukemia stem cells (LSCs) or pre-LSCs to arsenic trioxide (ATO), represent the main problems following treatment with ATO alone in patients with MDS. To solve these problems, a chemosensitization agent can be applied to increase the susceptibility of these cells to ATO. Curcumin (CUR), which possesses a wide range of anticancer activities, is a commonly used chemosensitization agent for various types of tumors, including hematopoietic malignancies. In the present study, we investigated the cytotoxic effects and potential mechanisms in MDS-SKM-1 and leukemia stem-like KG1a cells treated with CUR and ATO alone or in combination. CUR and ATO exhibited growth inhibition detected by MTT assays and apoptosis analyzed by Annexin V/PI analyses in both SKM-1 and KG1a cells. Apoptosis of SKM-1 and KG1a cells determined by Annexin V/PI was significantly enhanced in the combination groups compared with the groups treated with either agent alone. Further evaluation was performed by western blotting for two hallmark markers of apoptosis, caspase-3 and cleaved-PARP. Co-treatment of the cells with CUR and ATO resulted in significant synergistic effects. In SKM-1 and KG1a cells, 31 and 13 proteins analyzed by protein array assays were modulated, respectively. Notably, survivin protein expression levels were downregulated in both cell lines treated with CUR alone and in combination with ATO, particularly in the latter case. Susceptibility to apoptosis was significantly increased in SKM-1 and KG1a cells treated with siRNA-survivin and ATO. These results suggested that CUR increased the sensitivity of SKM-1 and KG1a cells to ATO by downregulating the expression of survivin.
Collapse
Affiliation(s)
- Yingjian Zeng
- Department of Hematology, Affiliated Jiangmen Traditional Chinese Medical Hospital of Jinan University, Jiangmen, Guangdong 529000, P.R. China
| | - Guangyang Weng
- Deparment of Hematology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518000, P.R. China
| | - Jiaxin Fan
- Department of Hematology, Affiliated Jiangmen Traditional Chinese Medical Hospital of Jinan University, Jiangmen, Guangdong 529000, P.R. China
| | - Zhangqiu Li
- Department of Hematology, Affiliated Jiangmen Traditional Chinese Medical Hospital of Jinan University, Jiangmen, Guangdong 529000, P.R. China
| | - Jianwei Wu
- Department of Hematology, Affiliated Jiangmen Traditional Chinese Medical Hospital of Jinan University, Jiangmen, Guangdong 529000, P.R. China
| | - Yuanming Li
- Department of Hematology, Affiliated Jiangmen Traditional Chinese Medical Hospital of Jinan University, Jiangmen, Guangdong 529000, P.R. China
| | - Rong Zheng
- Department of Hematology, Affiliated Jiangmen Traditional Chinese Medical Hospital of Jinan University, Jiangmen, Guangdong 529000, P.R. China
| | - Pingfang Xia
- Department of Hematology, Affiliated Jiangmen Traditional Chinese Medical Hospital of Jinan University, Jiangmen, Guangdong 529000, P.R. China
| | - Kunyuan Guo
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, P.R. China
| |
Collapse
|
21
|
Evidence that high-migration drug-surviving MOLT4 leukemia cells exhibit cancer stem cell-like properties. Int J Oncol 2016; 49:343-51. [PMID: 27210806 DOI: 10.3892/ijo.2016.3526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/28/2016] [Indexed: 11/05/2022] Open
Abstract
Leukemia represents a spectrum of hematological malignancies threatening human health. Resistance to treatments and metastasis of leukemia are the main causes of death in patients. Leukemia stem cells (LSCs) are the initiating cells of leukemia as well as the main source of drug resistance, invasion and metastasis. Consequently, eliminating LSCs is a prerequisite to eradicate leukemia. Preliminary studies in our laboratory have shown that chemokines and their related receptors play an important role in the drug resistance and metastasis of leukemic cells. In this study, we obtained high migration drug-surviving (short term) MOLT4 cells (hMDSCs-MOLT4) with treatment of doxorubicin (DOX) after Transwell assay. Then we detected stem cell-associated molecular markers on hMDSCs-MOLT4 cells and the parental MOLT4 cells by FCM, QPCR, western blotting, H&E staining and immunohisto-chemistry experimental techniques in vitro and in vivo. Moreover, we explored its impact on drug resistance and tumor formation. Then we found that compared with the parental MOLT4 cells, the mRNA expression levels of stem cell-related factors Sox2, Oct4, C-myc, Klf4, Nanog, Bmi-1, CXCR4 are increased in hMDSCs-MOLT4 cells, together with the protein expression levels of Sox2, Oct4, Klf4, Nanog, CXCR4 and CD34. Our results indicated that hMDSCs-MOLT4 cells exhibited strong drug resistance and certain cancer stem cell-like characteristics. It is the first indication that the targeting stemness factors such as Sox2, Oct4, Klf4, Nanog and CXCR4 may represent plausible options for eliminating T-ALL stem-like cells. The present findings shed light on the relationship between drug-tolerant leukemic cells and cancer stem cells.
Collapse
|
22
|
Nicolay JP, Schneider S, Gaiser T, Felcht M, Klemke CD. Differential expression of cancer stem cell markers in cutaneous and systemic lymphoma. Exp Dermatol 2016; 25:561-3. [DOI: 10.1111/exd.12951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Jan P. Nicolay
- Department of Dermatology, Venereology and Allergy; University Medical Center Mannheim; Ruprecht-Karls-University of Heidelberg; Mannheim Germany
- Department of Immunogenetics; German Cancer Research Center; Heidelberg Germany
| | - Sven Schneider
- Institute for Clinical Chemistry; University Medical Center Mannheim; Ruprecht-Karls-University of Heidelberg; Mannheim Germany
| | - Timo Gaiser
- Institute for Pathology; University Medical Center Mannheim; Ruprecht-Karls-University of Heidelberg; Mannheim Germany
| | - Moritz Felcht
- Department of Dermatology, Venereology and Allergy; University Medical Center Mannheim; Ruprecht-Karls-University of Heidelberg; Mannheim Germany
| | - Claus-Detlev Klemke
- Department of Dermatology, Venereology and Allergy; University Medical Center Mannheim; Ruprecht-Karls-University of Heidelberg; Mannheim Germany
- Hautklinik, Städtisches Klinikum Karlsruhe; Karlsruhe Germany
| |
Collapse
|
23
|
Gerber JM, Zeidner JF, Morse S, Blackford AL, Perkins B, Yanagisawa B, Zhang H, Morsberger L, Karp J, Ning Y, Gocke CD, Rosner GL, Smith BD, Jones RJ. Association of acute myeloid leukemia's most immature phenotype with risk groups and outcomes. Haematologica 2016; 101:607-16. [PMID: 26819054 DOI: 10.3324/haematol.2015.135194] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/22/2016] [Indexed: 11/09/2022] Open
Abstract
The precise phenotype and biology of acute myeloid leukemia stem cells remain controversial, in part because the "gold standard" immunodeficient mouse engraftment assay fails in a significant fraction of patients and identifies multiple cell-types in others. We sought to analyze the clinical utility of a novel assay for putative leukemia stem cells in a large prospective cohort. The leukemic clone's most primitive hematopoietic cellular phenotype was prospectively identified in 109 newly-diagnosed acute myeloid leukemia patients, and analyzed against clinical risk groups and outcomes. Most (80/109) patients harbored CD34(+)CD38(-) leukemia cells. The CD34(+)CD38(-) leukemia cells in 47 of the 80 patients displayed intermediate aldehyde dehydrogenase expression, while normal CD34(+)CD38(-) hematopoietic stem cells expressed high levels of aldehyde dehydrogenase. In the other 33/80 patients, the CD34(+)CD38(-) leukemia cells exhibited high aldehyde dehydrogenase activity, and most (28/33, 85%) harbored poor-risk cytogenetics or FMS-like tyrosine kinase 3 internal tandem translocations. No CD34(+) leukemia cells could be detected in 28/109 patients, including 14/21 patients with nucleophosmin-1 mutations and 6/7 acute promyelocytic leukemia patients. The patients with CD34(+)CD38(-) leukemia cells with high aldehyde dehydrogenase activity manifested a significantly lower complete remission rate, as well as poorer event-free and overall survivals. The leukemic clone's most immature phenotype was heterogeneous with respect to CD34, CD38, and ALDH expression, but correlated with acute myeloid leukemia risk groups and outcomes. The strong clinical correlations suggest that the most immature phenotype detectable in the leukemia might serve as a biomarker for "clinically-relevant" leukemia stem cells. ClinicalTrials.gov: NCT01349972.
Collapse
Affiliation(s)
| | - Joshua F Zeidner
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Sarah Morse
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Amanda L Blackford
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | | | - Breann Yanagisawa
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Hao Zhang
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Laura Morsberger
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Judith Karp
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Yi Ning
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher D Gocke
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Gary L Rosner
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - B Douglas Smith
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| | - Richard J Jones
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
24
|
Kersten B, Valkering M, Wouters R, van Amerongen R, Hanekamp D, Kwidama Z, Valk P, Ossenkoppele G, Zeijlemaker W, Kaspers G, Cloos J, Schuurhuis GJ. CD45RA, a specific marker for leukaemia stem cell sub-populations in acute myeloid leukaemia. Br J Haematol 2016; 173:219-35. [PMID: 26814163 DOI: 10.1111/bjh.13941] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 11/20/2015] [Indexed: 12/24/2022]
Abstract
Chemotherapy resistant leukaemic stem cells (LSC) are thought to be responsible for relapses after therapy in acute myeloid leukaemia (AML). Flow cytometry can discriminate CD34(+) CD38(-) LSC and normal haematopoietic stem cells (HSC) by using aberrant expression of markers and scatter properties. However, not all LSC can be identified using currently available markers, so new markers are needed. CD45RA is expressed on leukaemic cells in the majority of AML patients. We investigated the potency of CD45RA to specifically identify LSC and HSC and improve LSC quantification. Compared to our best other markers (CLL-1, also termed CLEC12A, CD33 and CD123), CD45RA was the most reliable marker. Patients with high percentages (>90%) of CD45RA on CD34(+) CD38(-) LSC have 1·69-fold higher scatter values compared to HSC (P < 0·001), indicating a more mature CD34(+) CD38(-) phenotype. Patients with low (<10%) or intermediate (10-90%) CD45RA expression on LSC showed no significant differences to HSC (1·12- and 1·15-fold higher, P = 0·31 and P = 0·44, respectively). CD45RA-positive LSC tended to represent more favourable cytogenetic/molecular markers. In conclusion, CD45RA contributes to more accurate LSC detection and is recommended for inclusion in stem cell tracking panels. CD45RA may contribute to define new LSC-specific therapies and to monitor effects of anti-LSC treatment.
Collapse
Affiliation(s)
- Bas Kersten
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Matthijs Valkering
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Rolf Wouters
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Rosa van Amerongen
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Diana Hanekamp
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Zinia Kwidama
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Peter Valk
- Department of Haematology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Gert Ossenkoppele
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | | | - Gertjan Kaspers
- Department of Paediatric Oncology/Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Jacqueline Cloos
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands.,Department of Paediatric Oncology/Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Gerrit J Schuurhuis
- Department of Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| |
Collapse
|
25
|
Khan M, DiNardo CD. Great expectations in acute myeloid leukemia. Future Oncol 2016; 12:289-92. [PMID: 26768493 DOI: 10.2217/fon.15.308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Maliha Khan
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA
| | - Courtney D DiNardo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, USA
| |
Collapse
|
26
|
Lang F, Wojcik B, Rieger MA. Stem Cell Hierarchy and Clonal Evolution in Acute Lymphoblastic Leukemia. Stem Cells Int 2015; 2015:137164. [PMID: 26236346 PMCID: PMC4506911 DOI: 10.1155/2015/137164] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 01/15/2023] Open
Abstract
Cancer is characterized by a remarkable intertumoral, intratumoral, and cellular heterogeneity that might be explained by the cancer stem cell (CSC) and/or the clonal evolution models. CSCs have the ability to generate all different cells of a tumor and to reinitiate the disease after remission. In the clonal evolution model, a consecutive accumulation of mutations starting in a single cell results in competitive growth of subclones with divergent fitness in either a linear or a branching succession. Acute lymphoblastic leukemia (ALL) is a highly malignant cancer of the lymphoid system in the bone marrow with a dismal prognosis after relapse. However, stabile phenotypes and functional data of CSCs in ALL, the so-called leukemia-initiating cells (LICs), are highly controversial and the question remains whether there is evidence for their existence. This review discusses the concepts of CSCs and clonal evolution in respect to LICs mainly in B-ALL and sheds light onto the technical controversies in LIC isolation and evaluation. These aspects are important for the development of strategies to eradicate cells with LIC capacity. Common properties of LICs within different subclones need to be defined for future ALL diagnostics, treatment, and disease monitoring to improve the patients' outcome in ALL.
Collapse
Affiliation(s)
- Fabian Lang
- Department of Hematology/Oncology, Goethe University Hospital, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Bartosch Wojcik
- Department of Hematology/Oncology, Goethe University Hospital, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- LOEWE Center for Cell and Gene Therapy Frankfurt, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Michael A. Rieger
- Department of Hematology/Oncology, Goethe University Hospital, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- LOEWE Center for Cell and Gene Therapy Frankfurt, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
27
|
Abstract
Parthenolide (PTL) has shown great promise as a novel anti-leukemia agent as it selectively eliminates acute myeloid leukemia (AML) blast cells and leukemia stem cells (LSCs) while sparing normal hematopoietic cells. This success has not yet translated to the clinical setting because PTL is rapidly cleared from blood due to its hydrophobicity. To increase the aqueous solubility of PTL, we previously developed micelles formed from predominantly hydrophobic amphiphilic diblock copolymers of poly(styrene-alt-maleic anhydride)-b-poly(styrene) (e.g., PSMA100-b-PS258) that exhibit robust PTL loading (75%efficiency, 11% w/w capacity) and release PTL over 24 h. Here, PTL-loaded PSMA-b-PS micelles were thoroughly characterized in vitro for PTL delivery to MV4-11 AML cells. Additionally, the mechanisms governing micelle-mediated cytotoxicity were examined in comparison to free PTL. PSMA-b-PS micelles were taken up by MV4-11 cells as evidenced by transmission electron microscopy and flow cytometry. Specifically, MV4-11 cells relied on clathrin-mediated endocytosis, rather than caveolae-mediated endocytosis and macropinocytosis. In addition, PTL-loaded PSMA-b-PS micelles exhibited a dose-dependent cytotoxicity towards AML cells and were capable of reducing cell viability by 75% at 10 μM PTL, while unloaded micelles were nontoxic. At 10 μM PTL, the cytotoxicity of PTL-loaded micelles increased gradually over 24 h while free PTL achieved maximal cytotoxicity between 2 and 4 h, demonstrating micelle-mediated delivery of PTL to AML cells and stability of the drug-loaded micelle even in the presence of cells. Both free PTL and PTL-loaded micelles induced NF-κB inhibition at 10 μM PTL doses, demonstrating some mechanistic similarities in cytotoxicity. However, free PTL relied more heavily on exofacial free thiol interactions to induce cytotoxicity than PTL-loaded micelles; free PTL cytotoxicity was reduced by over twofold when cell surface free thiols were depleted, where PTL-loaded micelle doses were unaffected by cell surface thiol modulation. The physical properties, stability, and efficacy of PTL-loaded PSMA-b-PS micelles support further development of a leukemia therapeutic with greater bioavailability and the potential to eliminate LSCs.
Collapse
|
28
|
Hoang VT, Buss EC, Wang W, Hoffmann I, Raffel S, Zepeda-Moreno A, Baran N, Wuchter P, Eckstein V, Trumpp A, Jauch A, Ho AD, Lutz C. The rarity of ALDH(+) cells is the key to separation of normal versus leukemia stem cells by ALDH activity in AML patients. Int J Cancer 2015; 137:525-36. [PMID: 25545165 PMCID: PMC4755039 DOI: 10.1002/ijc.29410] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 12/03/2014] [Indexed: 12/26/2022]
Abstract
To understand the precise disease driving mechanisms in acute myeloid leukemia (AML), comparison of patient matched hematopoietic stem cells (HSC) and leukemia stem cells (LSC) is essential. In this analysis, we have examined the value of aldehyde dehydrogenase (ALDH) activity in combination with CD34 expression for the separation of HSC from LSC in 104 patients with de novo AML. The majority of AML patients (80 out of 104) had low percentages of cells with high ALDH activity (ALDH(+) cells; <1.9%; ALDH-rare AML), whereas 24 patients had relatively numerous ALDH(+) cells (≥1.9%; ALDH-numerous AML). In patients with ALDH-rare AML, normal HSC could be separated by their CD34(+) ALDH(+) phenotype, whereas LSC were exclusively detected among CD34(+) ALDH(-) cells. For patients with ALDH-numerous AML, the CD34(+) ALDH(+) subset consisted mainly of LSC and separation from HSC was not feasible. Functional analyses further showed that ALDH(+) cells from ALDH-numerous AML were quiescent, refractory to ARA-C treatment and capable of leukemic engraftment in a xenogenic mouse transplantation model. Clinically, resistance to chemotherapy and poor long-term outcome were also characteristic for patients with ALDH-numerous AML providing an additional risk-stratification tool. The difference in spectrum and relevance of ALDH activity in the putative LSC populations demonstrates, in addition to phenotypic and genetic, also functional heterogeneity of leukemic cells and suggests divergent roles for ALDH activity in normal HSC versus LSC. By acknowledging these differences our study provides a new and useful tool for prospective identification of AML cases in which separation of HSC from LSC is possible.
Collapse
Affiliation(s)
- Van T Hoang
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Eike C Buss
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Wenwen Wang
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Isabel Hoffmann
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Simon Raffel
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGMBH), Heidelberg, Germany.,Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Abraham Zepeda-Moreno
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany.,Child and Youth Cancer Research Institute, University Center for Health Sciences, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Natalia Baran
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Patrick Wuchter
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Volker Eckstein
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGMBH), Heidelberg, Germany.,Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Anthony D Ho
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Christoph Lutz
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
29
|
Establishment of a humanized APL model via the transplantation of PML-RARA-transduced human common myeloid progenitors into immunodeficient mice. PLoS One 2014; 9:e111082. [PMID: 25369030 PMCID: PMC4219701 DOI: 10.1371/journal.pone.0111082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 09/24/2014] [Indexed: 11/19/2022] Open
Abstract
Recent advances in cancer biology have revealed that many malignancies possess a hierarchal system, and leukemic stem cells (LSC) or leukemia-initiating cells (LIC) appear to be obligatory for disease progression. Acute promyelocytic leukemia (APL), a subtype of acute myeloid leukemia characterized by the formation of a PML-RARα fusion protein, leads to the accumulation of abnormal promyelocytes. In order to understand the precise mechanisms involved in human APL leukemogenesis, we established a humanized in vivo APL model involving retroviral transduction of PML-RARA into CD34+ hematopoietic cells from human cord blood and transplantation of these cells into immunodeficient mice. The leukemia well recapitulated human APL, consisting of leukemic cells with abundant azurophilic abnormal granules in the cytoplasm, which expressed CD13, CD33 and CD117, but not HLA-DR and CD34, were clustered in the same category as human APL samples in the gene expression analysis, and demonstrated sensitivity to ATRA. As seen in human APL, the induced APL cells showed a low transplantation efficiency in the secondary recipients, which was also exhibited in the transplantations that were carried out using the sorted CD34− fraction. In order to analyze the mechanisms underlying APL initiation and development, fractionated human cord blood was transduced with PML-RARA. Common myeloid progenitors (CMP) from CD34+/CD38+ cells developed APL. These findings demonstrate that CMP are a target fraction for PML-RARA in APL, whereas the resultant CD34− APL cells may share the ability to maintain the tumor.
Collapse
|
30
|
Wouters R, Cucchi D, Kaspers GJL, Schuurhuis GJ, Cloos J. Relevance of leukemic stem cells in acute myeloid leukemia: heterogeneity and influence on disease monitoring, prognosis and treatment design. Expert Rev Hematol 2014; 7:791-805. [PMID: 25242511 DOI: 10.1586/17474086.2014.959921] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Acute myeloid leukemia is a bone marrow disease characterized by a block in differentiation of the myeloid lineage with a concomitant uncontrolled high proliferation rate. Development of acute myeloid leukemia from stem cells with specific founder mutations, leads to an oligoclonal disease that progresses into a very heterogeneous leukemia at diagnosis. Measurement of leukemic stem cell load and characterization of these cells are essential for prediction of relapse and target identification, respectively. Prediction of relapse by monitoring the disease during minimal residual disease detection is challenged by clonal shifts during therapy. To overcome this, characterization of the potential relapse-initiating cells is required using both flow cytometry and molecular analysis since leukemic stem cells can be targeted both on extracellular features and on stem-cell specific signal transduction pathways.
Collapse
Affiliation(s)
- Rolf Wouters
- Departments of Pediatric Oncology/Hematology and Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
31
|
Terwijn M, Zeijlemaker W, Kelder A, Rutten AP, Snel AN, Scholten WJ, Pabst T, Verhoef G, Löwenberg B, Zweegman S, Ossenkoppele GJ, Schuurhuis GJ. Leukemic stem cell frequency: a strong biomarker for clinical outcome in acute myeloid leukemia. PLoS One 2014; 9:e107587. [PMID: 25244440 PMCID: PMC4171508 DOI: 10.1371/journal.pone.0107587] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/11/2014] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Treatment failure in acute myeloid leukemia is probably caused by the presence of leukemia initiating cells, also referred to as leukemic stem cells, at diagnosis and their persistence after therapy. Specific identification of leukemia stem cells and their discrimination from normal hematopoietic stem cells would greatly contribute to risk stratification and could predict possible relapses. RESULTS For identification of leukemic stem cells, we developed flow cytometric methods using leukemic stem cell associated markers and newly-defined (light scatter) aberrancies. The nature of the putative leukemic stem cells and normal hematopoietic stem cells, present in the same patient's bone marrow, was demonstrated in eight patients by the presence or absence of molecular aberrancies and/or leukemic engraftment in NOD-SCID IL-2Rγ-/- mice. At diagnosis (n=88), the frequency of the thus defined neoplastic part of CD34+CD38- putative stem cell compartment had a strong prognostic impact, while the neoplastic parts of the CD34+CD38+ and CD34- putative stem cell compartments had no prognostic impact at all. After different courses of therapy, higher percentages of neoplastic CD34+CD38- cells in complete remission strongly correlated with shorter patient survival (n=91). Moreover, combining neoplastic CD34+CD38- frequencies with frequencies of minimal residual disease cells (n=91), which reflect the total neoplastic burden, revealed four patient groups with different survival. CONCLUSION AND PERSPECTIVE Discrimination between putative leukemia stem cells and normal hematopoietic stem cells in this large-scale study allowed to demonstrate the clinical importance of putative CD34+CD38- leukemia stem cells in AML. Moreover, it offers new opportunities for the development of therapies directed against leukemia stem cells, that would spare normal hematopoietic stem cells, and, moreover, enables in vivo and ex vivo screening for potential efficacy and toxicity of new therapies.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/metabolism
- Adolescent
- Adult
- Animals
- Antigens, CD34/metabolism
- Biomarkers
- Cell Count
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Immunophenotyping
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Middle Aged
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Prognosis
- Young Adult
Collapse
Affiliation(s)
- Monique Terwijn
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Angèle Kelder
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Arjo P. Rutten
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Alexander N. Snel
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Thomas Pabst
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gregor Verhoef
- Department of Hematology, University Hospital Leuven, Leuven, Belgium
| | - Bob Löwenberg
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Gert J. Ossenkoppele
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Gerrit J. Schuurhuis
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
32
|
Ossenkoppele GJ, Schuurhuis GJ. MRD in AML: It is time to change the definition of remission. Best Pract Res Clin Haematol 2014; 27:265-71. [DOI: 10.1016/j.beha.2014.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
33
|
Humme D, Lukowsky A, Gierisch M, Haider A, Vandersee S, Assaf C, Sterry W, Möbs M, Beyer M. T-cell receptor gene rearrangement analysis of sequential biopsies in cutaneous T-cell lymphomas with the Biomed-2 PCR reveals transient T-cell clones in addition to the tumor clone. Exp Dermatol 2014; 23:504-8. [DOI: 10.1111/exd.12453] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Daniel Humme
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Ansgar Lukowsky
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Maria Gierisch
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Ahmed Haider
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Staffan Vandersee
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Chalid Assaf
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
- HELIOS Klinikum Krefeld; Krefeld Germany
| | - Wolfram Sterry
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Markus Möbs
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - Marc Beyer
- Department of Dermatology and Allergy; Charité - Universitätsmedizin Berlin; Berlin Germany
| |
Collapse
|
34
|
Dorritie KA, Redner RL, Johnson DE. STAT transcription factors in normal and cancer stem cells. Adv Biol Regul 2014; 56:30-44. [PMID: 24931719 DOI: 10.1016/j.jbior.2014.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 05/21/2014] [Accepted: 05/22/2014] [Indexed: 01/02/2023]
Abstract
Signal transducer and activator of transcription proteins (STATs) play vital roles in the regulation of cellular proliferation and survival in normal hematopoietic cells, including hematopoietic stem cells. However, aberrant activation of STATs is commonly observed in a number of hematologic malignancies, and recent studies indicate that targeting of STATs may have therapeutic benefit in these diseases. Additional studies have provided greater understanding of the cells responsible for leukemia initiation, referred to as leukemia stem cells. Emerging evidence indicates that STATs are important in maintaining leukemia stem cells and represent a promising target for eradication of this dangerous cell population. Here we summarize what is known about normal hematopoietic stem cells and the origin of leukemic stem cells. We further describe the roles of STAT proteins in these cell populations, as well as current progress toward the development of novel agents and strategies for targeting the STAT proteins.
Collapse
Affiliation(s)
- Kathleen A Dorritie
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
| | - Robert L Redner
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Daniel E Johnson
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
35
|
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
|
36
|
Flavaglines target primitive leukemia cells and enhance anti-leukemia drug activity. Leukemia 2014; 28:1960-8. [PMID: 24577530 DOI: 10.1038/leu.2014.93] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 02/19/2014] [Accepted: 02/21/2014] [Indexed: 01/28/2023]
Abstract
Identification of agents that target human leukemia stem cells is an important consideration for the development of new therapies. The present study demonstrates that rocaglamide and silvestrol, closely related natural products from the flavagline class of compounds, are able to preferentially kill functionally defined leukemia stem cells, while sparing normal stem and progenitor cells. In addition to efficacy as single agents, flavaglines sensitize leukemia cells to several anticancer compounds, including front-line chemotherapeutic drugs used to treat leukemia patients. Mechanistic studies indicate that flavaglines strongly inhibit protein synthesis, leading to the reduction of short-lived antiapoptotic proteins. Notably though, treatment with flavaglines, alone or in combination with other drugs, yields a much stronger cytotoxic activity toward leukemia cells than the translational inhibitor temsirolimus. These results indicate that the underlying cell death mechanism of flavaglines is more complex than simply inhibiting general protein translation. Global gene expression profiling and cell biological assays identified Myc inhibition and the disruption of mitochondrial integrity to be features of flavaglines, which we propose contribute to their efficacy in targeting leukemia cells. Taken together, these findings indicate that rocaglamide and silvestrol are distinct from clinically available translational inhibitors and represent promising candidates for the treatment of leukemia.
Collapse
|
37
|
Zhang L, Ren X, Cheng Y, Liu X, Allen JE, Zhang Y, Yuan Y, Huang SY, Yang W, Berg A, Webb BS, Connor J, Liu CG, Lu Z, El-Deiry WS, Yang JM. The NFκB inhibitor, SN50, induces differentiation of glioma stem cells and suppresses their oncogenic phenotype. Cancer Biol Ther 2014; 15:602-11. [PMID: 24557012 DOI: 10.4161/cbt.28158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The malignant phenotype of glioblastoma multiforme (GBM) is believed to be largely driven by glioma stem-like cells (GSCs), and targeting GSCs is now considered a promising new approach to treatment of this devastating disease. Here, we show that SN50, a cell-permeable peptide inhibitor of NFκB, induced robust differentiation of human GSCs, causing loss of their oncogenic potential. We observed that following treatment of GSCs with SN50, their differentiated progeny cells showed significant decreases in their capability to form neuro-spheres and to invade in vitro and a reduction in their tumorigenicity in mouse xenograft models, but had increased sensitivity to the chemotherapeutic drug temozolomide and to radiation treatment. These results suggest that blocking the NFκB pathway may be explored as a useful mean to induce differentiation of GSCs, and provide another supportive evidence for the promise of differentiation therapy in treatment of malignant brain tumors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacology; College of Pharmaceutical Sciences; Soochow University; Suzhou, PR China; Department of Pharmacology; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Xingcong Ren
- Department of Pharmacology; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Yan Cheng
- Department of Pharmacology; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Xiuping Liu
- Department of Experimental Therapeutics; MD Anderson Cancer Center; Houston, TX USA
| | - Joshua E Allen
- Department of Medicine; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Yi Zhang
- Department of Pharmacology; College of Pharmaceutical Sciences; Soochow University; Suzhou, PR China
| | - Yunsheng Yuan
- Department of Pharmacology; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Siu-Yuan Huang
- Department of Pharmacology; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Weiwei Yang
- Department of Neuro-Oncology; MD Anderson Cancer Center; Houston, TX USA
| | - Arthur Berg
- Department of Public Health Sciences; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Becky S Webb
- Department of Neurosurgery; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - James Connor
- Department of Neurosurgery; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Chang-Gong Liu
- Department of Experimental Therapeutics; MD Anderson Cancer Center; Houston, TX USA
| | - Zhimin Lu
- Department of Neuro-Oncology; MD Anderson Cancer Center; Houston, TX USA
| | - Wafik S El-Deiry
- Department of Medicine; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| | - Jin-Ming Yang
- Department of Pharmacology; College of Pharmaceutical Sciences; Soochow University; Suzhou, PR China; Department of Pharmacology; The Penn State Hershey Cancer Institute; The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center; Hershey, PA USA
| |
Collapse
|
38
|
Identifying arsenic trioxide (ATO) functions in leukemia cells by using time series gene expression profiles. Gene 2014; 535:312-7. [DOI: 10.1016/j.gene.2013.10.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/18/2013] [Accepted: 10/30/2013] [Indexed: 01/30/2023]
|
39
|
Differential requirement for wild-type Flt3 in leukemia initiation among mouse models of human leukemia. Exp Hematol 2013; 42:192-203.e1. [PMID: 24269847 DOI: 10.1016/j.exphem.2013.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 10/08/2013] [Accepted: 11/12/2013] [Indexed: 11/24/2022]
Abstract
FLT3 is one of the most frequently mutated genes in acute leukemias. However, the role in leukemogenesis of wild-type (wt) FLT3, which is highly expressed in many hematologic malignancies, is unclear. We show here that in mouse models established by retroviral transduction of leukemic fusion proteins, deletion of Flt3 strongly inhibits MLL-ENL and to lesser extent p210(BCR-ABL)-induced leukemogenesis, but has no effect in MLL-AF9 or AML1-ETO9a models. Flt3 acts at the level of leukemic stem cells (LSCs), as a fraction of LSCs in MLL-ENL, but not in MLL-AF9-induced leukemia, expressed Flt3 in vivo, and Flt3 expression on LSCs was associated with leukemia development in this model. Furthermore, efficiency of MLL-ENL, but not of MLL-AF9-induced leukemia induction was significantly enhanced after transduction of Flt3(+) compared to Flt3(-) wt myeloid progenitors. However, Flt3 is not required for immortalization of bone marrow cells in vitro by MLL-ENL and does not affect colony formation by MLL-ENL LSCs in vitro, suggesting that in vitro models do not reflect the in vivo biology of MLL-ENL leukemia with respect to Flt3 requirement. We conclude that wt Flt3 plays a role in leukemia initiation in vivo, which is, however, not universal.
Collapse
|
40
|
Abstract
Aging is a degenerative process resulting in compromised tissue maintenance and increased susceptibility to diseases, such as cancer. Recent advancements support the notion that aging is a highly regulated process governed by evolutionarily conserved pathways. In mammals, tissue-specific adult stem cells (ASCs) persist throughout the lifetime to maintain and repair tissues. While reduced ASC self-renewal is thought to contribute to compromised tissue maintenance, increased self-renewal of cancer stem cells (CSCs) may lead to tumorigenesis. It is speculated that genetic regulators of aging, such as sirtuins, are likely to impinge upon the ASC compartments to regulate tissue maintenance and tumorigenesis. In this review, we discuss the emerging evidence linking sirtuins to normal and malignant ASC self-renewal, tissue maintenance, and tumorigenesis.
Collapse
|
41
|
Zhang J, Seet CS, Sun C, Li J, You D, Volk A, Breslin P, Li X, Wei W, Qian Z, Zeleznik-Le NJ, Zhang Z, Zhang J. p27kip1 maintains a subset of leukemia stem cells in the quiescent state in murine MLL-leukemia. Mol Oncol 2013; 7:1069-82. [PMID: 23988911 DOI: 10.1016/j.molonc.2013.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 07/22/2013] [Accepted: 07/31/2013] [Indexed: 12/14/2022] Open
Abstract
MLL (mixed-lineage leukemia)-fusion genes induce the development of leukemia through deregulation of normal MLL target genes, such as HOXA9 and MEIS1. Both HOXA9 and MEIS1 are required for MLL-fusion gene-induced leukemogenesis. Co-expression of HOXA9 and MEIS1 induces acute myeloid leukemia (AML) similar to that seen in mice in which MLL-fusion genes are over-expressed. p27(kip1) (p27 hereafter), a negative regulator of the cell cycle, has also been defined as an MLL target, the expression of which is up-regulated in MLL leukemic cells (LCs). To investigate whether p27 plays a role in the pathogenesis of MLL-leukemia, we examined the effects of p27 deletion (p27(-/-)) on MLL-AF9 (MA9)-induced murine AML development. HOXA9/MEIS1 (H/M)-induced, p27 wild-type (p27(+/+)) and p27(-/-) AML were studied in parallel as controls. We found that LCs from both MA9-AML and H/M-AML can be separated into three fractions, a CD117(-)CD11b(hi) differentiated fraction as well as CD117(+)CD11b(hi) and CD117(+)CD11b(lo), two less differentiated fractions. The CD117(+)CD11b(lo) fraction, comprising only 1-3% of total LCs, expresses higher levels of early hematopoietic progenitor markers but lower levels of mature myeloid cell markers compared to other populations of LCs. p27 is expressed and is required for maintaining the quiescent and drug-resistant states of the CD117(+)CD11b(lo) fraction of MA9-LCs but not of H/M-LCs. p27 deletion significantly compromises the leukemogenic capacity of CD117(+)CD11b(lo) MA9-LCs by reducing the frequency of leukemic stem cells (LSCs) but does not do so in H/M-LCs. In addition, we found that p27 is highly expressed and required for cell cycle arrest in the CD117(-)CD11b(hi) fraction in both types of LCs. Furthermore, we found that c-Myc expression is required for maintaining LCs in an undifferentiated state independently of proliferation. We concluded that p27 represses the proliferation of LCs, which is specifically required for maintaining the quiescent and drug-resistant states of a small subset of MA9-LSCs in collaboration with the differentiation blockage function of c-Myc.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Biology, College of Life and Environment Science, Shanghai Normal University, 100 Guilin Road, Shanghai 200234, PR China; Oncology Institute, Cardinal Bernardin Cancer Center and Department of Pathology, Loyola University Chicago, Maywood, IL 60153, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mariani SA, Calabretta B. Leukemia stem cells: Old concepts and new perspectives. Mol Aspects Med 2013; 39:102-9. [PMID: 23820118 DOI: 10.1016/j.mam.2013.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 06/21/2013] [Indexed: 10/26/2022]
Abstract
Myeloid leukemias are heterogeneous malignancies in morphology, immunophenotype, genetic and epigenetic alterations, and response to therapy. This heterogeneity is thought to depend on the accumulation of secondary mutations enhancing proliferation/survival and/or blocking differentiation in a small subset of leukemia-initiating cells capable of self-renewal. This model of clonal evolution is based on xenotransplantation studies demonstrating that leukemia can be initiated and maintained in immunodeficient mice by a small subset of purified leukemic cells immunophenotypically similar to normal hematopoietic stem cells and is known as the leukemia stem cell model. Since its original formulation, many studies have validated the main conclusion of this model. However, recent data from xenotransplantation studies in more severely immunodeficient mice suggest that imunophenotype and behavior of leukemic stem cells is more heterogeneous and "plastic" than originally thought. We will discuss here the evolution of the leukemia stem cell model and its impact for the therapy of patients with myeloid malignancies.
Collapse
Affiliation(s)
- Samanta A Mariani
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Bruno Calabretta
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, United States.
| |
Collapse
|
43
|
Bell-Horwath TR, Vadukoot AK, Thowfeik FS, Li G, Wunderlich M, Mulloy JC, Merino EJ. Novel ROS-activated agents utilize a tethered amine to selectively target acute myeloid leukemia. Bioorg Med Chem Lett 2013; 23:2951-4. [PMID: 23578690 DOI: 10.1016/j.bmcl.2013.03.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/05/2013] [Accepted: 03/12/2013] [Indexed: 11/18/2022]
Abstract
This study explores the possible use of reactive oxygen-activated DNA modifying agents against acute myeloid leukemia (AML). A key amine on the lead agent was investigated via cytotoxicity assays and was found necessary for potency. The two best compounds were screened via the NCI-60 cell panel. These two compounds had potency between 200 and 800nM against many of the leukemia cancer cell types. Subsequent experiments explored activity against a transformed AML model that mimics the molecular signatures identified in primary AML patient samples. A lead compound had an IC50 of 760nM against this AML cell line as well as a therapeutic index of 7.7±3 between the transformed AML model cell line and non-cancerous human CD34+ blood stem/progenitor cells (UCB). The selectivity was much greater than the mainstays of AML treatment: doxorubicin and cytarabine. This manuscript demonstrates that this novel type of agent may be useful against AML.
Collapse
Affiliation(s)
- Tiffany R Bell-Horwath
- Department of Chemistry, University of Cincinnati, Cincinnati, OH 45221-0172, United States
| | | | | | | | | | | | | |
Collapse
|
44
|
Shman TV, Movchan LV, Aleinikova OV. Frequencies of immature CD34 + CD38 - and CD34 + CD38-CD19 + blasts correlate with minimal residual disease level in pediatric B-cell precursor acute lymphoblastic leukemia. Leuk Lymphoma 2013; 54:2560-2. [PMID: 23432723 DOI: 10.3109/10428194.2013.778404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Tatsiana V Shman
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology , Minsk , Belarus
| | | | | |
Collapse
|
45
|
Kurata M, Suzuki S, Abe S, Onishi I, Kitagawa M. Bone marrow cell death and proliferation: Controlling mechanisms in normal and leukemic state. World J Hematol 2013; 2:1-5. [DOI: 10.5315/wjh.v2.i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Bone marrow cell death and proliferation are regulated by multiple factors including genetic and epigenetic alterations of hematopoietic cells, crosstalk of hematopoietic cells with bone marrow mesenchymal cells through direct cell-cell interaction or cytokine/chemokine production, vascularity of the bone marrow, and interactions of sympathetic nerve system with hematopoiesis. Cell proliferation usually predominates over cell death in neoplastic processes such as leukemia and myeloproliferative neoplasms, while apoptotic processes also have a significant role in the pathogenesis of myelodysplastic syndromes. Recently, hematopoietic stem cells (HSCs) and leukemia stem cells (LSCs) have been identified and their characters on self renewal process, differentiation, cell dynamics and drug resistance have been implicated. Although most leukemia cells are initially sensitive to chemo- or radiotherapy, LSCs are resistant and considered to be the basis for disease relapse after initial response. HSCs and LSCs may use similar interactions with bone marrow microenvironment. However, bone marrow microenvironment called niche should influence the normal as well as malignant hematopoiesis in different manners. Recent studies have expanded the number of cell types constituting bone marrow niche and made the issue more complex. Since the majority of excellent and contributing studies on bone marrow niches have been performed in animal models, niches in human tissues are beginning to be localized and characterized. In this article, we summarize the relation of hematopoietic cells with niches and hope to point a hint to the novel strategy for treatment of malignant proliferation of hematopoietic cells.
Collapse
|
46
|
Abstract
As acute myeloid leukemia (AML) xenograft models improve, the potential for using them to evaluate novel therapeutic strategies becomes more appealing. Currently, there is little information on using standard chemotherapy regimens in AML xenografts. Here we have characterized the immunodeficient mouse response to combined Ara-C (cytarabine) and doxorubicin treatment. We observed significant toxicity associated with doxorubicin that required optimization of the route of injection as well as the maximum-tolerated dose for immunodeficient strains. Mice treated with an optimized 5-day induction protocol showed transient weight loss, short-term reduction of peripheral blood cell and platelet counts, and slight anemia. Considerable cytotoxicity was observed in the bone marrow (BM), with primitive LSK cells having a significant survival advantage relative to more mature cells, consistent with the idea of chemotherapy targeting actively growing cells. Treated leukemic mice demonstrated reduced disease burden and increased survival, demonstrating efficacy. AML cells showed significantly increased sensitivity to doxorubicin-containing therapy compared with murine BM cells. Although early treatment could result in some cures, mice with significant leukemia grafts were not cured by using induction therapy alone. Overall, the data show that this model system is useful for the evaluation of novel chemotherapies in combination with standard induction therapy.
Collapse
|
47
|
Huang SD, Yuan Y, Tang H, Liu XH, Fu CG, Cheng HZ, Bi JW, Yu YW, Gong DJ, Zhang W, Chen J, Xu ZY. Tumor cells positive and negative for the common cancer stem cell markers are capable of initiating tumor growth and generating both progenies. PLoS One 2013; 8:e54579. [PMID: 23349932 PMCID: PMC3549952 DOI: 10.1371/journal.pone.0054579] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 12/13/2012] [Indexed: 01/06/2023] Open
Abstract
The cancer stem cell (CSC) model depicts that tumors are hierarchically organized and maintained by CSCs lying at the apex. CSCs have been “identified” in a variety of tumors through the tumor-forming assay, in which tumor cells distinguished by a certain cell surface marker (known as a CSC marker) were separately transplanted into immunodeficient mice. In such assays, tumor cells positive but not negative for the CSC marker (hereby defined as CSC+ and CSC− cells, respectively) have the ability of tumor-forming and generating both progenies. However, here we show that CSC+ and CSC− cells exhibit similar proliferation in the native states. Using a cell tracing method, we demonstrate that CSC− cells exhibit similar tumorigenesis and proliferation as CSC+ cells when they were co-transplanted into immunodeficient mice. Through serial single-cell derived subline construction, we further demonstrated that CSC+ and CSC− cells from CSC marker expressing tumors could invariably generate both progenies, and their characteristics are maintained among different generations irrespective of the origins (CSC+-derived or CSC−-derived). These findings demonstrate that tumorigenic cells cannot be distinguished by common CSC markers alone and we propose that cautions should be taken when using these markers independently to identify cancer stem cells due to the phenotypic plasticity of tumor cells.
Collapse
Affiliation(s)
- Sheng-Dong Huang
- Institute of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, PR China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Dobrowolska H, Gill KZ, Serban G, Ivan E, Li Q, Qiao P, Suciu-Foca N, Savage D, Alobeid B, Bhagat G, Colovai AI. Expression of immune inhibitory receptor ILT3 in acute myeloid leukemia with monocytic differentiation. CYTOMETRY PART B-CLINICAL CYTOMETRY 2012; 84:21-9. [PMID: 23027709 DOI: 10.1002/cyto.b.21050] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 09/04/2012] [Accepted: 09/19/2012] [Indexed: 11/12/2022]
Abstract
BACKGROUND The diagnosis of AML with monocytic differentiation is limited by the lack of highly sensitive and specific monocytic markers. Immunoglobulin-like transcript 3 (ILT3) is an immune inhibitory receptor expressed by myelomonocytic cells and at high levels by tolerogenic dendritic cells. METHODS Using flow cytometry, we analyzed the expression of ILT3 in 37 patients with AML and 20 patients with no detectable disease. RESULTS We showed that ILT3 was expressed in all cases of AML displaying monocytic differentiation (FAB M4/M5; N = 18), but not in AML M1/M2 and M3 (N = 19; P < 0.0001). Co-expression of ILT3 and immature cell markers, such as CD34 and CD117, was observed in monoblastic leukemia. ILT3 expression was preserved after treatment in M4/M5 patients with refractory or relapsed disease. ILT3 expression was associated with the presence of cytogenetic abnormalities linked to an intermediate prognosis (P = 0.001). Rare CD45dimCD34+CD117+ILT3+ cells were identified in noninvolved bone marrow, suggesting that ILT3 expression is acquired at an early stage by normal myelomonocytic precursors. CONCLUSIONS ILT3 is a highly sensitive and specific marker which distinguishes AML with monocytic differentiation from other types of AML. Testing of ILT3 expression should be incorporated into the initial diagnostic work-up and monitoring of patients with AML.
Collapse
Affiliation(s)
- Hanna Dobrowolska
- Department of Pathology and Cell Biology, Columbia University Medical Center and New York Presbyterian Hospital, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Xia J, Chen C, Chen Z, Miele L, Sarkar FH, Wang Z. Targeting pancreatic cancer stem cells for cancer therapy. Biochim Biophys Acta Rev Cancer 2012; 1826:385-99. [PMID: 22728049 DOI: 10.1016/j.bbcan.2012.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 04/14/2012] [Accepted: 06/13/2012] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer (PC) is the fourth most frequent cause of cancer death in the United States. Emerging evidence suggests that pancreatic cancer stem cells (CSCs) play a crucial role in the development and progression of PC. Recently, there is increasing evidence showing that chemopreventive agents commonly known as nutraceuticals could target and eliminate CSCs that have been proposed as the root of the tumor progression, which could be partly due to attenuating cell signaling pathways involved in CSCs. Therefore, targeting pancreatic CSCs by nutraceuticals for the prevention of tumor progression and treatment of PC may lead to the development of novel strategy for achieving better treatment outcome of PC patients. In this review article, we will summarize the most recent advances in the pancreatic CSC field, with particular emphasis on nutraceuticals that target CSCs, for fighting this deadly disease.
Collapse
Affiliation(s)
- Jun Xia
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Anhui, People's Republic of China
| | | | | | | | | | | |
Collapse
|
50
|
Hutchinson SA, Tooke-Locke E, Wang J, Tsai S, Katz T, Trede NS. Tbl3 regulates cell cycle length during zebrafish development. Dev Biol 2012; 368:261-72. [PMID: 22659140 DOI: 10.1016/j.ydbio.2012.05.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Revised: 05/21/2012] [Accepted: 05/21/2012] [Indexed: 02/07/2023]
Abstract
The regulation of cell cycle rate is essential for the correct timing of proliferation and differentiation during development. Changes to cell cycle rate can have profound effects on the size, shape and cell types of a developing organ. We previously identified a zebrafish mutant ceylon (cey) that has a severe reduction in T cells and hematopoietic stem/progenitor cells (HSPCs). Here we find that the cey phenotype is due to absence of the gene transducin (beta)-like 3 (tbl3). The tbl3 homolog in yeast regulates the cell cycle by maintaining rRNA levels and preventing p53-induced cell death. Zebrafish tbl3 is maternally expressed, but later in development its expression is restricted to specific tissues. Tissues expressing tbl3 are severely reduced in cey mutants, including HSPCs, the retina, exocrine pancreas, intestine, and jaw cartilage. Specification of these tissues is normal, suggesting the reduced size is due to a reduced number of differentiated cells. Tbl3 MO injection into either wild-type or p53-/- mutant embryos phenocopies cey, indicating that loss of tbl3 causes specific defects in cey. Progression of both hematopoietic and retinal development is delayed beginning at 3 day post fertilization due to a slowing of the cell cycle. In contrast to yeast, reduction of Tbl3 causes a slowing of the cell cycle without a corresponding increase in p53 induced cell death. These data suggest that tbl3 plays a tissue-specific role regulating cell cycle rate during development.
Collapse
Affiliation(s)
- Sarah A Hutchinson
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | |
Collapse
|