1
|
Chen Y. Dissecting L-glutamine metabolism in acute myeloid leukemia: single-cell insights and therapeutic implications. J Transl Med 2024; 22:1002. [PMID: 39506790 PMCID: PMC11539756 DOI: 10.1186/s12967-024-05779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a rapidly progressing blood cancer. The prognosis of AML can be challenging, emphasizing the need for ongoing research and innovative approaches to improve outcomes in individuals affected by this formidable hematologic malignancy. METHODS In this study, we used single-cell RNA sequencing (scRNA-seq) from AML patients to investigate the impact of L-glutamine metabolism-related genes on disease progression. RESULTS Our analysis revealed increased glutamine-related activity in CD34 + pre-B cells, suggesting a potential regulatory role in tumorigenesis and AML progression. Furthermore, intercellular communication analysis revealed a significant signaling pathway involving macrophage migration inhibitory factor signaling through CD74 + CD44 within CD34 + pre-B cells, which transmit signals to pre-dendritic cells and monocytes. Ligands for this pathway were predominantly expressed in stromal cells, naïve T cells, and CD34 + pre-B cells. CD74, the pertinent receptor, was predominantly detected in a variety of cellular components, including stromal cells, pre-dendritic cells, plasmacytoid dendritic cells, and hematopoietic progenitors. The study's results provide insights into the possible interplay among these cell types and their collective contribution to the pathogenesis of AML. Moreover, we identified 10 genes associated with AML prognosis, including CCL5, CD52, CFD, FABP5, LGALS1, NUCB2, PSAP, S100A4, SPINK2, and VCAN. Among these, CCL5 and CD52 have been implicated in AML progression and are potential therapeutic targets. CONCLUSIONS This thorough examination of AML biology significantly deepens our grasp of the disease and presents pivotal information that could guide the creation of innovative treatment strategies for AML patients.
Collapse
Affiliation(s)
- Yanli Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
2
|
Ding L, Peng L, Huang K, Qu S, Li D, Yao J, Yang F, Zhu H, Zhao S. Single cell transcriptomics reveals dysregulated immnue homeostasis in different stages in HPV-induced cutaneous squamous cell carcinoma. Exp Dermatol 2024; 33:e15178. [PMID: 39385326 DOI: 10.1111/exd.15178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/04/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024]
Abstract
In order to explore the huge impact of impaired immnue homeostasis on the occurrence and development of cutaneous squamous cell carcinoma (cSCC), and investigate characterization of the cellular components and their changes which is crucial to understanding the pathologic process of HPV-induced cSCC, we diagnosed and followed up on a very rare HPV-induced cSCC patient who progressed at a very fast rate and transferred to death quickly. We performed single-cell RNA sequencing (scRNA-seq) of 11 379 cells from the skin tissues of this patient with four different skin statuses after HPV infection. Immunofluorescence experiments were used for validation. scRNA-seq identified that CD52+ HLA-DOA- macrophages only existed in paracancerous cutaneous squamous cell carcinoma (pc-cSCC) and cSCC tissue. Besides, immune cells including CD8+ exhausted T cells and CD4+ regulatory T cells as well as matrix cells like MMP1+, and MMP11+ fibroblasts were gradually increased. Meanwhile, COMP+ ASPN+ fibroblasts gradually decreased. Cell interaction analysis revealed enhancement in interactions between monocytes/macrophages, fibroblasts and tumour-specific keratinocytes. scRNA-seq was performed in HPV-induced cSCC for the first time, to explore the correlation between infection and tumour. It is the first time to study the development of tumours from different stages of infection in HPV-induced cSCC. In this study, the tumour itself and the tumour microenvironment were both analysed and explored. And it was validated in clinical samples from different patients. Our findings reveal the dynamic immnue homeostasis from normal skin to cSCC tissue, this alteration might drive HPV-induced cSCC.
Collapse
Affiliation(s)
- Liqing Ding
- The Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Lanyuan Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kai Huang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shunlin Qu
- Pathophysiology Department, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Dongjie Li
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
| | | | | | - Honglin Zhu
- The Department of Rheumatology and Immunology, Xiangya Hospital of Central South University, Changsha, Hunan, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Valent P, Sadovnik I, Peter B, Ivanov D, Schulenburg A, Hadzijusufovic E, Willmann M, Rülicke T, Herrmann H, Rabitsch W, Karlic H, Gleixner KV, Sperr WR, Hoermann G, Dahlhoff M, Pfeilstöcker M, Keil F, Lion T, Grunt TW. Vienna Cancer Stem Cell Club (VCSCC): 20 year jubilee and future perspectives. Expert Rev Hematol 2023; 16:659-670. [PMID: 37493441 DOI: 10.1080/17474086.2023.2232545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/29/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION The Vienna Cancer Stem Cell Club (VCSCC) was launched by a group of scientists in Vienna in 2002. AREAS COVERED Major aims of the VCSCC are to support research on cancer stem cells (CSC) in hematopoietic malignancies and to translate CSC-related markers and targets into clinical application. A primary focus of research in the VCSCC is the leukemic stem cell (LSC). Between 2013 and 2021, members of the VCSCC established a special research program on myeloproliferative neoplasms and since 2008, members of the VCSCC run the Ludwig Boltzmann Institute for Hematology and Oncology. In all these years, the VCSCC provided a robust intellectual platform for translational hematology and LSC research in Vienna. Furthermore, the VCSCC interacts with several national and international study groups and societies in the field. Representatives of the VCSCC also organized a number of international meetings and conferences on neoplastic stem cells, including LSC, in the past 15 years, and contributed to the definition and classification of CSC/LSC and related pre-malignant and malignant conditions. EXPERT OPINION The VCSCC will continue to advance the field and to develop LSC-detecting and LSC-eradicating concepts through which diagnosis, prognostication, and therapy of blood cancer patients should improve.
Collapse
Affiliation(s)
- Peter Valent
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Irina Sadovnik
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Barbara Peter
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Daniel Ivanov
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Axel Schulenburg
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Stem Cell Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Emir Hadzijusufovic
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Department for Companion Animals and Horses, University Clinic for Small Animals, Internal Medicine Small Animals, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Michael Willmann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department for Companion Animals and Horses, University Clinic for Small Animals, Internal Medicine Small Animals, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Rülicke
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Institute of in vivo and in vitro Models, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Harald Herrmann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Werner Rabitsch
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Stem Cell Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Heidrun Karlic
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Karoline V Gleixner
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang R Sperr
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - Maik Dahlhoff
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Institute of in vivo and in vitro Models, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Michael Pfeilstöcker
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Third Medical Department for Hematology and Oncology, Hanusch Hospital, Vienna, Austria
| | - Felix Keil
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Third Medical Department for Hematology and Oncology, Hanusch Hospital, Vienna, Austria
| | - Thomas Lion
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- St.Anna Children´s Cancer Research Institute (CCRI), Vienna, Austria
| | - Thomas W Grunt
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Division of Clinical Oncology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Morris JA, Caragine C, Daniloski Z, Domingo J, Barry T, Lu L, Davis K, Ziosi M, Glinos DA, Hao S, Mimitou EP, Smibert P, Roeder K, Katsevich E, Lappalainen T, Sanjana NE. Discovery of target genes and pathways at GWAS loci by pooled single-cell CRISPR screens. Science 2023; 380:eadh7699. [PMID: 37141313 PMCID: PMC10518238 DOI: 10.1126/science.adh7699] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/20/2023] [Indexed: 05/06/2023]
Abstract
Most variants associated with complex traits and diseases identified by genome-wide association studies (GWAS) map to noncoding regions of the genome with unknown effects. Using ancestrally diverse, biobank-scale GWAS data, massively parallel CRISPR screens, and single-cell transcriptomic and proteomic sequencing, we discovered 124 cis-target genes of 91 noncoding blood trait GWAS loci. Using precise variant insertion through base editing, we connected specific variants with gene expression changes. We also identified trans-effect networks of noncoding loci when cis target genes encoded transcription factors or microRNAs. Networks were themselves enriched for GWAS variants and demonstrated polygenic contributions to complex traits. This platform enables massively parallel characterization of the target genes and mechanisms of human noncoding variants in both cis and trans.
Collapse
Affiliation(s)
- John A. Morris
- New York Genome Center, New York, NY, 10013, USA
- Department of Biology, New York University, New York, NY, 10003, USA
| | | | - Zharko Daniloski
- New York Genome Center, New York, NY, 10013, USA
- Department of Biology, New York University, New York, NY, 10003, USA
| | | | - Timothy Barry
- Department of Statistics and Data Science, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Lu Lu
- New York Genome Center, New York, NY, 10013, USA
| | - Kyrie Davis
- New York Genome Center, New York, NY, 10013, USA
| | | | | | - Stephanie Hao
- Technology Innovation Lab, New York Genome Center, New York, NY, 10013, USA
| | - Eleni P. Mimitou
- Technology Innovation Lab, New York Genome Center, New York, NY, 10013, USA
| | - Peter Smibert
- Technology Innovation Lab, New York Genome Center, New York, NY, 10013, USA
| | - Kathryn Roeder
- Department of Statistics and Data Science, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Eugene Katsevich
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tuuli Lappalainen
- New York Genome Center, New York, NY, 10013, USA
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, 171 65 Solna, Stockholm, Sweden
| | - Neville E. Sanjana
- New York Genome Center, New York, NY, 10013, USA
- Department of Biology, New York University, New York, NY, 10003, USA
| |
Collapse
|
5
|
Single-cell analysis reveals the chemotherapy-induced cellular reprogramming and novel therapeutic targets in relapsed/refractory acute myeloid leukemia. Leukemia 2023; 37:308-325. [PMID: 36543880 PMCID: PMC9898038 DOI: 10.1038/s41375-022-01789-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Chemoresistance and relapse are the leading cause of AML-related deaths. Utilizing single-cell RNA sequencing (scRNA-seq), we dissected the cellular states of bone marrow samples from primary refractory or short-term relapsed AML patients and defined the transcriptional intratumoral heterogeneity. We found that compared to proliferating stem/progenitor-like cells (PSPs), a subpopulation of quiescent stem-like cells (QSCs) were involved in the chemoresistance and poor outcomes of AML. By performing longitudinal scRNA-seq analyses, we demonstrated that PSPs were reprogrammed to obtain a QSC-like expression pattern during chemotherapy in refractory AML patients, characterized by the upregulation of CD52 and LGALS1 expression. Flow cytometric analysis further confirmed that the preexisting CD99+CD49d+CD52+Galectin-1+ (QSCs) cells at diagnosis were associated with chemoresistance, and these cells were further enriched in the residual AML cells of refractory patients. Interaction of CD52-SIGLEC10 between QSCs and monocytes may contribute to immune evading and poor outcomes. Furthermore, we identified that LGALS1 was a promising target for chemoresistant AML, and LGALS1 inhibitor could help eliminate QSCs and enhance the chemotherapy in patient-derived primary AML cells, cell lines, and AML xenograft models. Our results will facilitate a better understanding of the AML chemoresistance mechanism and the development of novel therapeutic strategies for relapsed/refractory AML patients.
Collapse
|
6
|
Li F, Cai J, Liu J, Yu SC, Zhang X, Su Y, Gao L. Construction of a solid Cox model for AML patients based on multiomics bioinformatic analysis. Front Oncol 2022; 12:925615. [PMID: 36033493 PMCID: PMC9399435 DOI: 10.3389/fonc.2022.925615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematological malignancy. The bone marrow (BM) microenvironment in AML plays an important role in leukemogenesis, drug resistance and leukemia relapse. In this study, we aimed to identify reliable immune-related biomarkers for AML prognosis by multiomics analysis. We obtained expression profiles from The Cancer Genome Atlas (TCGA) database and constructed a LASSO-Cox regression model to predict the prognosis of AML using multiomics bioinformatic analysis data. This was followed by independent validation of the model in the GSE106291 (n=251) data set and mutated genes in clinical samples for predicting overall survival (OS). Molecular docking was performed to predict the most optimal ligands to the genes in prognostic model. The single-cell RNA sequence dataset GSE116256 was used to clarify the expression of the hub genes in different immune cell types. According to their significant differences in immune gene signatures and survival trends, we concluded that the immune infiltration-lacking subtype (IL type) is associated with better prognosis than the immune infiltration-rich subtype (IR type). Using the LASSO model, we built a classifier based on 5 hub genes to predict the prognosis of AML (risk score = -0.086×ADAMTS3 + 0.180×CD52 + 0.472×CLCN5 - 0.356×HAL + 0.368×ICAM3). In summary, we constructed a prognostic model of AML using integrated multiomics bioinformatic analysis that could serve as a therapeutic classifier.
Collapse
Affiliation(s)
- Fu Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jiao Cai
- Department of Hematology and Hematopoietic Stem Cell Transplantation Centre, The General Hospital of Western Theater Command, Chengdu, China
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jia Liu
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shi-cang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yi Su
- Department of Hematology and Hematopoietic Stem Cell Transplantation Centre, The General Hospital of Western Theater Command, Chengdu, China
- *Correspondence: Lei Gao, ; Yi Su,
| | - Lei Gao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- *Correspondence: Lei Gao, ; Yi Su,
| |
Collapse
|
7
|
Guess T, Potts CR, Bhat P, Cartailler JA, Brooks A, Holt C, Yenamandra A, Wheeler FC, Savona MR, Cartailler JP, Ferrell PB. Distinct Patterns of Clonal Evolution Drive Myelodysplastic Syndrome Progression to Secondary Acute Myeloid Leukemia. Blood Cancer Discov 2022; 3:316-329. [PMID: 35522837 PMCID: PMC9610896 DOI: 10.1158/2643-3230.bcd-21-0128] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/22/2022] [Accepted: 05/04/2022] [Indexed: 11/16/2022] Open
Abstract
Clonal evolution in myelodysplastic syndrome (MDS) can result in clinical progression and secondary acute myeloid leukemia (sAML). To dissect changes in clonal architecture associated with this progression, we performed single-cell genotyping of paired MDS and sAML samples from 18 patients. Analysis of single-cell genotypes revealed patient-specific clonal evolution and enabled the assessment of single-cell mutational cooccurrence. We discovered that changes in clonal architecture proceed via distinct patterns, classified as static or dynamic, with dynamic clonal architectures having a more proliferative phenotype by blast count fold change. Proteogenomic analysis of a subset of patients confirmed that pathogenic mutations were primarily confined to primitive and mature myeloid cells, though we also identify rare but present mutations in lymphocyte subsets. Single-cell transcriptomic analysis of paired sample sets further identified gene sets and signaling pathways involved in two cases of progression. Together, these data define serial changes in the MDS clonal landscape with clinical and therapeutic implications. SIGNIFICANCE Precise clonal trajectories in MDS progression are made possible by single-cell genomic sequencing. Here we use this technology to uncover the patterns of clonal architecture and clonal evolution that drive the transformation to secondary AML. We further define the phenotypic and transcriptional changes of disease progression at the single-cell level. See related article by Menssen et al., p. 330 (31). See related commentary by Romine and van Galen, p. 270. This article is highlighted in the In This Issue feature, p. 265.
Collapse
Affiliation(s)
- Tiffany Guess
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee.,Department of Pathology, Microbiology, and Immunology, VUMC, Nashville, Tennessee
| | - Chad R. Potts
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
| | - Pawan Bhat
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Justin A. Cartailler
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
| | - Austin Brooks
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
| | - Clinton Holt
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ashwini Yenamandra
- Department of Pathology, Microbiology, and Immunology, VUMC, Nashville, Tennessee
| | - Ferrin C. Wheeler
- Department of Pathology, Microbiology, and Immunology, VUMC, Nashville, Tennessee
| | - Michael R. Savona
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee.,Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Jean-Philippe Cartailler
- Creative Data Solutions Shared Resource, Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee
| | - P. Brent Ferrell
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee.,Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Vanderbilt-Ingram Cancer Center, Nashville, Tennessee.,Corresponding Author: P. Brent Ferrell Jr, Vanderbilt University Medical Center, 777 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232. Phone: 615-875-8619; E-mail:
| |
Collapse
|
8
|
CD52 is a novel target for the treatment of FLT3-ITD-mutated myeloid leukemia. Cell Death Discov 2021; 7:121. [PMID: 34035227 PMCID: PMC8149417 DOI: 10.1038/s41420-021-00446-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/22/2021] [Accepted: 03/09/2021] [Indexed: 12/01/2022] Open
Abstract
Internal tandem duplication (ITD) of FMS-like tyrosine kinase 3 (FLT3) confers poor prognosis and is found in approximately 25% of cases of acute myeloid leukemia (AML). Although FLT3 inhibitors have shown clinical benefit in patients with AML harboring FLT3-ITD, the therapeutic effect is limited. Here, to explore alternative therapeutics, we established a cellular model of monoallelic FLT3ITD/WT cells using the CRISPR-Cas9 system in a human myeloid leukemia cell line, K562. cDNA microarray analysis revealed elevated CD52 expression in K562–FLT3ITD/WT cells compared to K562–FLT3WT/WT cells, an observation that was further confirmed by quantitative real-time-PCR and flow cytometric analyses. The elevated expression of CD52 in K562–FLT3ITD/WT cells was decreased in wild-type FLT3 (FLT3-WT) knock-in K562–FLT3ITD/WT cells. In K562–FLT3ITD/WT cells, a STAT5 inhibitor, pimozide, downregulated CD52 protein expression while an AKT inhibitor, afuresertib, did not affect CD52 expression. Notably, an anti-CD52 antibody, alemtuzumab, induced significant antibody-dependent cell-mediated cytotoxicity (ADCC) in K562-FLT3ITD/WT cells compared to K562–FLT3WT/WT cells. Furthermore, alemtuzumab significantly suppressed the xenograft tumor growth of K562–FLT3ITD/WT cells in severe combined immunodeficiency (SCID) mice. Taken together, our data suggested that genetically modified FLT3-ITD knock-in human myeloid leukemia K562 cells upregulated CD52 expression via activation of STAT5, and alemtuzumab showed an antitumor effect via induction of ADCC in K562–FLT3ITD/WT cells. Our findings may allow establishment of a new therapeutic option, alemtuzumab, to treat leukemia with the FLT3-ITD mutation.
Collapse
|
9
|
Delineation of target expression profiles in CD34+/CD38- and CD34+/CD38+ stem and progenitor cells in AML and CML. Blood Adv 2021; 4:5118-5132. [PMID: 33085758 DOI: 10.1182/bloodadvances.2020001742] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
In an attempt to identify novel markers and immunological targets in leukemic stem cells (LSCs) in acute myeloid leukemia (AML) and chronic myeloid leukemia (CML), we screened bone marrow (BM) samples from patients with AML (n = 274) or CML (n = 97) and controls (n = 288) for expression of cell membrane antigens on CD34+/CD38- and CD34+/CD38+ cells by multicolor flow cytometry. In addition, we established messenger RNA expression profiles in purified sorted CD34+/CD38- and CD34+/CD38+ cells using gene array and quantitative polymerase chain reaction. Aberrantly expressed markers were identified in all cohorts. In CML, CD34+/CD38- LSCs exhibited an almost invariable aberration profile, defined as CD25+/CD26+/CD56+/CD93+/IL-1RAP+. By contrast, in patients with AML, CD34+/CD38- cells variably expressed "aberrant" membrane antigens, including CD25 (48%), CD96 (40%), CD371 (CLL-1; 68%), and IL-1RAP (65%). With the exception of a subgroup of FLT3 internal tandem duplication-mutated patients, AML LSCs did not exhibit CD26. All other surface markers and target antigens detected on AML and/or CML LSCs, including CD33, CD44, CD47, CD52, CD105, CD114, CD117, CD133, CD135, CD184, and roundabout-4, were also found on normal BM stem cells. However, several of these surface targets, including CD25, CD33, and CD123, were expressed at higher levels on CD34+/CD38- LSCs compared with normal BM stem cells. Moreover, antibody-mediated immunological targeting through CD33 or CD52 resulted in LSC depletion in vitro and a substantially reduced LSC engraftment in NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice. Together, we have established surface marker and target expression profiles of AML LSCs and CML LSCs, which should facilitate LSC enrichment, diagnostic LSC phenotyping, and development of LSC-eradicating immunotherapies.
Collapse
|
10
|
Valent P, Bauer K, Sadovnik I, Smiljkovic D, Ivanov D, Herrmann H, Filik Y, Eisenwort G, Sperr WR, Rabitsch W. Cell-based and antibody-mediated immunotherapies directed against leukemic stem cells in acute myeloid leukemia: Perspectives and open issues. Stem Cells Transl Med 2020; 9:1331-1343. [PMID: 32657052 PMCID: PMC7581453 DOI: 10.1002/sctm.20-0147] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022] Open
Abstract
Despite new insights in molecular features of leukemic cells and the availability of novel treatment approaches and drugs, acute myeloid leukemia (AML) remains a major clinical challenge. In fact, many patients with AML relapse after standard therapy and eventually die from progressive disease. The basic concept of leukemic stem cells (LSC) has been coined with the goal to decipher clonal architectures in various leukemia-models and to develop curative drug therapies by eliminating LSC. Indeed, during the past few years, various immunotherapies have been tested in AML, and several of these therapies follow the strategy to eliminate relevant leukemic subclones by introducing LSC-targeting antibodies or LSC-targeting immune cells. These therapies include, among others, new generations of LSC-eliminating antibody-constructs, checkpoint-targeting antibodies, bi-specific antibodies, and CAR-T or CAR-NK cell-based strategies. However, responses are often limited and/or transient which may be due to LSC resistance. Indeed, AML LSC exhibit multiple forms of resistance against various drugs and immunotherapies. An additional problems are treatment-induced myelotoxicity and other side effects. The current article provides a short overview of immunological targets expressed on LSC in AML. Moreover, cell-based therapies and immunotherapies tested in AML are discussed. Finally, the article provides an overview about LSC resistance and strategies to overcome resistance.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Karin Bauer
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Dubravka Smiljkovic
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
| | - Daniel Ivanov
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
| | - Harald Herrmann
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
- Department of Radiation OncologyMedical University of ViennaViennaAustria
| | - Yüksel Filik
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Gregor Eisenwort
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Werner Rabitsch
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
- Department of Internal Medicine I, Stem Cell Transplantation UnitMedical University of ViennaViennaAustria
| |
Collapse
|
11
|
Zou Z, Ha Y, Liu S, Huang B. Identification of tumor-infiltrating immune cells and microenvironment-relevant genes in nasopharyngeal carcinoma based on gene expression profiling. Life Sci 2020; 263:118620. [PMID: 33096113 DOI: 10.1016/j.lfs.2020.118620] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/30/2020] [Accepted: 10/11/2020] [Indexed: 12/24/2022]
Abstract
AIMS The purpose of this study was to investigate the prognostic significance of tumor-infiltrating immune cells and microenvironment-relevant genes in nasopharyngeal carcinoma (NPC) and their correlations. MATERIALS AND METHODS The "xCell" algorithm was used to calculate the enrichment scores for 33 immune cells in the samples of GSE12452, GSE40290, GSE53819, GSE68799, and GSE102349. The difference of immune cells between NPC group and non-cancerous group and the prognostic value of the immune cells were analyzed. Besides, based on the Microenvironment scores, the differentially expressed genes (DEGs) between the high- and low-score groups were screened to identify the microenvironment-relevant hub genes. Furthermore, the DEGs were used to establish a risk score model for predicting progression-free survival (PFS) via LASSO penalized Cox regression. KEY FINDINGS The scores of B-cells and Memory B-cells of NPC were significantly lower than those of non-cancerous tissues, and they were positively associated with PFS. Moreover, 10 hub genes (PTPRC, CD19, CD79B, BTK, CD79A, SELL, MS4A1, CD38, CD52, and CD22) were identified and positively correlated with B-cells, Memory B-cells, and Microenvironment scores in GSE12452, GSE68799, and GSE102349. High expression levels of CD22, CD38, CD79B, MS4A1, SELL, and PTPRC were associated with longer PFS. Besides, a risk score model composed of DARC, IL33, IGHG1, and SLC6A8 was established with a good performance for PFS prediction. SIGNIFICANCE These results enhance our understanding of the composition and prognostic significance of tumor-infiltrating immune cells in NPC lesions, and provide potential targets for prognostication and immunotherapy for NPC patients.
Collapse
Affiliation(s)
- Zhenning Zou
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Yanping Ha
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Shuguang Liu
- Department of Pathology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bowan Huang
- Department of Anesthesiology, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
12
|
Tan Y, Wu Q, Zhou F. Targeting acute myeloid leukemia stem cells: Current therapies in development and potential strategies with new dimensions. Crit Rev Oncol Hematol 2020; 152:102993. [PMID: 32502928 DOI: 10.1016/j.critrevonc.2020.102993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
High relapse rate of acute myeloid leukemia (AML) is still a crucial problem despite considerable advances in anti-cancer therapies. One crucial cause of relapse is the existence of leukemia stem cells (LSCs) with self-renewal ability, which contribute to repeated treatment resistance and recurrence. Treatments targeting LSCs, especially in combination with existing chemotherapy regimens or hematopoietic stem cell transplantation might help achieve a higher complete remission rate and improve overall survival. Many novel agents of different therapeutic strategies that aim to modulate LSCs self-renewal, proliferation, apoptosis, and differentiation are under investigation. In this review, we summarize the latest advances of different therapies in development based on the biological characteristics of LSCs, with particular attention on natural products, synthetic compounds, antibody therapies, and adoptive cell therapies that promote the LSC eradication. We also explore the causes of AML recurrence and proposed potential strategies with new dimensions for targeting LSCs in the future.
Collapse
Affiliation(s)
- Yuxin Tan
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.
| |
Collapse
|
13
|
Mitchell K, Steidl U. Targeting Immunophenotypic Markers on Leukemic Stem Cells: How Lessons from Current Approaches and Advances in the Leukemia Stem Cell (LSC) Model Can Inform Better Strategies for Treating Acute Myeloid Leukemia (AML). Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036251. [PMID: 31451539 DOI: 10.1101/cshperspect.a036251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Therapies targeting cell-surface antigens in acute myeloid leukemia (AML) have been tested over the past 20 years with limited improvement in overall survival. Recent advances in the understanding of AML pathogenesis support therapeutic targeting of leukemia stem cells as the most promising avenue toward a cure. In this review, we provide an overview of the evolving leukemia stem cell (LSC) model, including evidence of the cell of origin, cellular and molecular disease architecture, and source of relapse in AML. In addition, we explore limitations of current targeted strategies utilized in AML and describe the various immunophenotypic antigens that have been proposed as LSC-directed therapeutic targets. We draw lessons from current approaches as well as from the (pre)-LSC model to suggest criteria that immunophenotypic targets should meet for more specific and effective elimination of disease-initiating clones, highlighting in detail a few targets that we suggest fit these criteria most completely.
Collapse
Affiliation(s)
- Kelly Mitchell
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, New York 10461, USA.,Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
14
|
Lin SY, Miao YR, Hu FF, Hu H, Zhang Q, Li Q, Chen Z, Guo AY. A 6-Membrane Protein Gene score for prognostic prediction of cytogenetically normal acute myeloid leukemia in multiple cohorts. J Cancer 2020; 11:251-259. [PMID: 31892991 PMCID: PMC6930412 DOI: 10.7150/jca.35382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Cytogenetically normal acute myeloid leukemia (CN-AML) is a large proportion of AMLs with diverse prognostic outcomes. Identifying membrane protein genes as prognostic factors to stratify CN-AML patients will be critical to improve their outcomes. Purpose: This study aims to identify prognostic factors to stratify CN-AML patients to choose better treatments and improve their outcomes. Methods: CN-AML data were from TCGA cohort (n = 79) and four GEO datasets. We identified independent prognostic genes by Cox regression and Kaplan-Meier methods, and constructed linear regression model using LASSO algorithm. The prediction error curve was calculated using R package “pec”. Results: Based on independent prognostic membrane genes, we constructed a regression model for CN-AML prognosis prediction: score = (0.0492 * CD52) - (0.0018 * CD96) + (0.0131 * EMP1) + (0.2058 * TSPAN2) + (0.0234 * STAB1) - (0.3658 * MBTPS1), which was named as MPG6 (6-Membrane Protein Gene) score. Tested in multiple CN-AML datasets, consistent results showed that CN-AML patients with high MPG6 score had poor survival, higher WBC count and shorter EFS. Comparing with other reported scoring models, the benchmark result of MPG6 achieved better association with survival in multiple cohorts. Moreover, by combining with other clinical indicators in CN-AML, MPG6 could improve the performance of survival prediction and serve as a robust prognostic factor. Conclusions: We identified the MPG6 score as a stable indicator with great potential for clinical application in risk stratification and outcome prediction in CN-AML.
Collapse
Affiliation(s)
- Sheng-Yan Lin
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ya-Ru Miao
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Fei-Fei Hu
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Hui Hu
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qiong Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qiubai Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhichao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - An-Yuan Guo
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
15
|
Xu ZJ, Gu Y, Wang CZ, Jin Y, Wen XM, Ma JC, Tang LJ, Mao ZW, Qian J, Lin J. The M2 macrophage marker CD206: a novel prognostic indicator for acute myeloid leukemia. Oncoimmunology 2019; 9:1683347. [PMID: 32002295 PMCID: PMC6959428 DOI: 10.1080/2162402x.2019.1683347] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 01/13/2023] Open
Abstract
Hematological malignancies possess a distinctive immunologic microenvironment compared with solid tumors. Here, using an established computational algorithm (CIBERSORT), we systematically analyzed the overall distribution of 22 tumor-infiltrating leukocyte (TIL) populations in more than 2000 bone marrow (BM) samples from 5 major hematological malignancies and healthy controls. Focusing on significantly altered TILs in acute myeloid leukemia (AML), we found that patients with AML exhibited increased frequencies of M2 macrophages, compared to either healthy controls or the other four malignancies. High infiltration of M2 macrophages was associated with poor outcome in AML. Further analysis revealed that CD206, a M2 marker gene, could faithfully reflect variation in M2 fractions and was more highly expressed in AML than normal controls. High CD206 expression predicted inferior overall survival (OS) and event-free survival (EFS) in two independent AML cohorts. Among 175 patients with intermediate-risk cytogenetics, the survival still differed greatly between low and high CD206 expressers (OS; P < .0001; 3-year rates, 56% v 32%; EFS; P < .001; 3-year rates, 47% v 25%). When analyzed in a meta-analysis, CD206 as a continuous variable showed superior predictive performance than classical prognosticators in AML (BAALC, ERG, EVI1, MN1, and WT1). In summary, M2 macrophages are preferentially enriched in AML. The M2 marker CD206 may serve as a new prognostic marker in AML.
Collapse
Affiliation(s)
- Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Yu Gu
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Cui-Zhu Wang
- Department of Oncology, Affiliated Haian Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Ye Jin
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Xiang-Mei Wen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Li-Juan Tang
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Zhen-Wei Mao
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| | - Jun Qian
- Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P.R. China.,Zhenjiang Clinical Research Center of Hematology, Zhenjiang, Jiangsu, P.R. China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang, Jiangsu, P.R. China
| |
Collapse
|
16
|
Valent P, Sadovnik I, Eisenwort G, Bauer K, Herrmann H, Gleixner KV, Schulenburg A, Rabitsch W, Sperr WR, Wolf D. Immunotherapy-Based Targeting and Elimination of Leukemic Stem Cells in AML and CML. Int J Mol Sci 2019; 20:E4233. [PMID: 31470642 PMCID: PMC6747233 DOI: 10.3390/ijms20174233] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/25/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022] Open
Abstract
The concept of leukemic stem cells (LSC) has been developed with the idea to explain the clonal hierarchies and architectures in leukemia, and the more or less curative anti-neoplastic effects of various targeted drugs. It is now widely accepted that curative therapies must have the potential to eliminate or completely suppress LSC, as only these cells can restore and propagate the malignancy for unlimited time periods. Since LSC represent a minor cell fraction in the leukemic clone, little is known about their properties and target expression profiles. Over the past few years, several cell-specific immunotherapy concepts have been developed, including new generations of cell-targeting antibodies, antibody-toxin conjugates, bispecific antibodies, and CAR-T cell-based strategies. Whereas such concepts have been translated and may improve outcomes of therapy in certain lymphoid neoplasms and a few other malignancies, only little is known about immunological targets that are clinically relevant and can be employed to establish such therapies in myeloid neoplasms. In the current article, we provide an overview of the immunologically relevant molecular targets expressed on LSC in patients with acute myeloid leukemia (AML) and chronic myeloid leukemia (CML). In addition, we discuss the current status of antibody-based therapies in these malignancies, their mode of action, and successful examples from the field.
Collapse
MESH Headings
- Acute Disease
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- CTLA-4 Antigen/metabolism
- Humans
- Immunologic Factors/therapeutic use
- Immunotherapy/methods
- Immunotherapy/trends
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/therapy
- Molecular Targeted Therapy/methods
- Molecular Targeted Therapy/trends
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria.
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gregor Eisenwort
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Karin Bauer
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Harald Herrmann
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria
- Department of Radiotherapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Karoline V Gleixner
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Axel Schulenburg
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria
- Division of Blood and Bone Marrow Transplantation, Department of Internal Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Werner Rabitsch
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria
- Division of Blood and Bone Marrow Transplantation, Department of Internal Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dominik Wolf
- Department of Internal Medicine V (Hematology & Oncology), Medical University of Innsbruck, 1090 Innsbruck, Austria
- Medical Clinic 3, Oncology, Hematology, Immunoncology & Rheumatology, University Clinic Bonn (UKB), 53127 Bonn, Germany
| |
Collapse
|
17
|
Houshmand M, Blanco TM, Circosta P, Yazdi N, Kazemi A, Saglio G, Zarif MN. Bone marrow microenvironment: The guardian of leukemia stem cells. World J Stem Cells 2019; 11:476-490. [PMID: 31523368 PMCID: PMC6716085 DOI: 10.4252/wjsc.v11.i8.476] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/13/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023] Open
Abstract
Bone marrow microenvironment (BMM) is the main sanctuary of leukemic stem cells (LSCs) and protects these cells against conventional therapies. However, it may open up an opportunity to target LSCs by breaking the close connection between LSCs and the BMM. The elimination of LSCs is of high importance, since they follow cancer stem cell theory as a part of this population. Based on cancer stem cell theory, a cell with stem cell-like features stands at the apex of the hierarchy and produces a heterogeneous population and governs the disease. Secretion of cytokines, chemokines, and extracellular vesicles, whether through autocrine or paracrine mechanisms by activation of downstream signaling pathways in LSCs, favors their persistence and makes the BMM less hospitable for normal stem cells. While all details about the interactions of the BMM and LSCs remain to be elucidated, some clinical trials have been designed to limit these reciprocal interactions to cure leukemia more effectively. In this review, we focus on chronic myeloid leukemia and acute myeloid leukemia LSCs and their milieu in the bone marrow, how to segregate them from the normal compartment, and finally the possible ways to eliminate these cells.
Collapse
Affiliation(s)
- Mohammad Houshmand
- Department of Clinical and Biological Sciences, University of Turin, Turin 10126, Italy
| | - Teresa Mortera Blanco
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm 14183, Sweden
| | - Paola Circosta
- Department of Clinical and Biological Sciences, University of Turin, Turin 10126, Italy
| | - Narjes Yazdi
- Department of Molecular Genetics, Tehran Medical Branch, Islamic Azad University, Tehran 1916893813, Iran
| | - Alireza Kazemi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin, Turin 10126, Italy
| | - Mahin Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran 146651157, Iran
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm 14183, Sweden
| |
Collapse
|
18
|
Detecting the stable point of therapeutic effect of chronic myeloid leukemia based on dynamic network biomarkers. BMC Bioinformatics 2019; 20:202. [PMID: 31074387 PMCID: PMC6509869 DOI: 10.1186/s12859-019-2738-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background Most researches of chronic myeloid leukemia (CML) are currently focused on the treatment methods, while there are relatively few researches on the progress of patients’ condition after drug treatment. Traditional biomarkers of disease can only distinguish normal state from disease state, and cannot recognize the pre-stable state after drug treatment. Results A therapeutic effect recognition strategy based on dynamic network biomarkers (DNB) is provided for CML patients’ gene expression data. With the DNB criteria, the DNB with 250 genes is selected and the therapeutic effect index (TEI) is constructed for the detection of individual disease. The pre-stable state before the disease condition becomes stable is 1 month. Through functional analysis for the DNB, some genes are confirmed as key genes to affect the progress of CML patients’ condition. Conclusions The results provide a certain theoretical direction and theoretical basis for medical personnel in the treatment of CML patients, and find new therapeutic targets in the future. The biomarkers of CML can help patients to be treated promptly and minimize drug resistance, treatment failure and relapse, which reduce the mortality of CML significantly. Electronic supplementary material The online version of this article (10.1186/s12859-019-2738-0) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
Eisenwort G, Sadovnik I, Schwaab J, Jawhar M, Keller A, Stefanzl G, Berger D, Blatt K, Hoermann G, Bilban M, Willmann M, Winding C, Sperr WR, Arock M, Rülicke T, Reiter A, Valent P. Identification of a leukemia-initiating stem cell in human mast cell leukemia. Leukemia 2019; 33:2673-2684. [PMID: 30953030 PMCID: PMC6839966 DOI: 10.1038/s41375-019-0460-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 03/17/2019] [Accepted: 03/18/2019] [Indexed: 12/23/2022]
Abstract
Mast cell leukemia (MCL) is a highly fatal malignancy characterized by devastating expansion of immature mast cells in various organs. Although considered a stem cell disease, little is known about MCL-propagating neoplastic stem cells. We here describe that leukemic stem cells (LSCs) in MCL reside within a CD34+/CD38− fraction of the clone. Whereas highly purified CD34+/CD38− cells engrafted NSGhSCF mice with fully manifesting MCL, no MCL was produced by CD34+/CD38+ progenitors or the bulk of KIT+/CD34− mast cells. CD34+/CD38− MCL cells invariably expressed CD13 and CD133, and often also IL-1RAP, but did not express CD25, CD26 or CLL-1. CD34+/CD38− MCL cells also displayed several surface targets, including CD33, which was homogenously expressed on MCL LSC in all cases, as well as the D816V mutant form of KIT. Whereas CD34+/CD38− cells were resistant against single drugs, exposure to combinations of CD33-targeting and KIT-targeting drugs resulted in LSC-depletion and markedly reduced engraftment in NSGhSCF mice. Together, MCL LSCs are CD34+/CD38− cells that express distinct profiles of markers and target antigens. Characterization of MCL LSCs should facilitate their purification and should support the development of LSC-eradicating curative treatment approaches in this fatal type of leukemia.
Collapse
Affiliation(s)
- Gregor Eisenwort
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
| | - Irina Sadovnik
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
| | - Juliana Schwaab
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Mohamad Jawhar
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Alexandra Keller
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
| | - Gabriele Stefanzl
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
| | - Daniela Berger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
| | - Katharina Blatt
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Willmann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090, Vienna, Austria.,Department for Companion Animals & Horses, Clinic for Internal Medicine and Infectious Diseases, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Christiana Winding
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Wolfgang R Sperr
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michel Arock
- Cellular and Molecular Oncology, LBPA CNRS UMR8113, Ecole Normale Supérieure de Cachan, 94230, Cachan, France
| | - Thomas Rülicke
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090, Vienna, Austria.,Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Andreas Reiter
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090, Vienna, Austria. .,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
20
|
Valent P, Hadzijusufovic E, Grunt T, Karlic H, Peter B, Herrmann H, Eisenwort G, Hoermann G, Schulenburg A, Willmann M, Hubmann R, Shehata M, Selzer E, Gleixner KV, Rülicke T, Sperr WR, Marian B, Pfeilstöcker M, Pehamberger H, Keil F, Jäger U, Zielinski C. Ludwig Boltzmann Cluster Oncology (LBC ONC): first 10 years and future perspectives. Wien Klin Wochenschr 2018; 130:517-529. [PMID: 30006759 PMCID: PMC6132878 DOI: 10.1007/s00508-018-1355-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/14/2018] [Indexed: 12/17/2022]
Abstract
In 2008 the Ludwig Boltzmann Cluster Oncology (LBC ONC) was established on the basis of two previous Ludwig Boltzmann Institutes working in the field of hematology and cancer research. The general aim of the LBC ONC is to improve treatment of hematopoietic neoplasms by eradicating cancer-initiating and disease-propagating cells, also known as leukemic stem cells (LSC) in the context of leukemia. In a first phase, the LBC ONC characterized the phenotype and molecular aberration profiles of LSC in various malignancies. The LSC phenotypes were established in acute and chronic myeloid leukemia, in acute lymphoblastic leukemia and in chronic lymphocytic leukemia. In addition, the concept of preleukemic (premalignant) neoplastic stem cells (pre-L-NSC) was coined by the LBC ONC and was tested in myelodysplastic syndromes and myeloproliferative neoplasms. Phenotypic characterization of LSC provided a solid basis for their purification and for the characterization of specific target expression profiles. In a second phase, molecular markers and targets were validated. This second phase is ongoing and should result in the development of new diagnostics parameters and novel, more effective, LSC-eradicating, treatment strategies; however, many issues still remain to be solved, such as sub-clonal evolution, LSC niche interactions, immunologic control of LSC, and LSC resistance. In the forthcoming years, the LBC ONC will concentrate on developing LSC-eradicating strategies, with special focus on LSC resistance, precision medicine and translation of LSC-eradicating concepts into clinical application.
Collapse
Affiliation(s)
- Peter Valent
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria. .,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria.
| | - Emir Hadzijusufovic
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria.,Department/Clinic for Companion Animals and Horses, Clinic for Small Animals, Clinical Unit of Internal Medicine, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Grunt
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Division of Clinical Oncology, Medical University of Vienna, Vienna, Austria
| | - Heidrun Karlic
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Hanusch Hospital, Vienna, Austria
| | - Barbara Peter
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Harald Herrmann
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Gregor Eisenwort
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Gregor Hoermann
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Axel Schulenburg
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Stem Cell Transplantation Unit, Medical University of Vienna, Vienna, Austria
| | - Michael Willmann
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Companion Animals and Horses, Clinic for Internal Medicine and Infectious Diseases, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Rainer Hubmann
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Medhat Shehata
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Edgar Selzer
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Karoline V Gleixner
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Thomas Rülicke
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Wolfgang R Sperr
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Brigitte Marian
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Michael Pfeilstöcker
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Hanusch Hospital, Vienna, Austria
| | - Hubert Pehamberger
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Felix Keil
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Hanusch Hospital, Vienna, Austria
| | - Ulrich Jäger
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Christoph Zielinski
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.,Department of Internal Medicine I, Division of Clinical Oncology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
21
|
Schürch CM. Therapeutic Antibodies for Myeloid Neoplasms-Current Developments and Future Directions. Front Oncol 2018; 8:152. [PMID: 29868474 PMCID: PMC5968093 DOI: 10.3389/fonc.2018.00152] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) such as antibody-drug conjugates, ligand-receptor antagonists, immune checkpoint inhibitors and bispecific T cell engagers have shown impressive efficacy in the treatment of multiple human cancers. Numerous therapeutic mAbs that have been developed for myeloid neoplasms, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), are currently investigated in clinical trials. Because AML and MDS originate from malignantly transformed hematopoietic stem/progenitor cells-the so-called leukemic stem cells (LSCs) that are highly resistant to most standard drugs-these malignancies frequently relapse and have a high disease-specific mortality. Therefore, combining standard chemotherapy with antileukemic mAbs that specifically target malignant blasts and particularly LSCs or utilizing mAbs that reinforce antileukemic host immunity holds great promise for improving patient outcomes. This review provides an overview of therapeutic mAbs for AML and MDS. Antibody targets, the molecular mechanisms of action, the efficacy in preclinical leukemia models, and the results of clinical trials are discussed. New developments and future studies of therapeutic mAbs in myeloid neoplasms will advance our understanding of the immunobiology of these diseases and enhance current therapeutic strategies.
Collapse
Affiliation(s)
- Christian M. Schürch
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
22
|
Blatt K, Menzl I, Eisenwort G, Cerny-Reiterer S, Herrmann H, Herndlhofer S, Stefanzl G, Sadovnik I, Berger D, Keller A, Hauswirth A, Hoermann G, Willmann M, Rülicke T, Sill H, Sperr WR, Mannhalter C, Melo JV, Jäger U, Sexl V, Valent P. Phenotyping and Target Expression Profiling of CD34 +/CD38 - and CD34 +/CD38 + Stem- and Progenitor cells in Acute Lymphoblastic Leukemia. Neoplasia 2018; 20:632-642. [PMID: 29772458 PMCID: PMC5994777 DOI: 10.1016/j.neo.2018.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022] Open
Abstract
Leukemic stem cells (LSCs) are an emerging target of curative anti-leukemia therapy. In acute lymphoblastic leukemia (ALL), LSCs frequently express CD34 and often lack CD38. However, little is known about markers and targets expressed in ALL LSCs. We have examined marker- and target expression profiles in CD34+/CD38− LSCs in patients with Ph+ ALL (n = 22) and Ph− ALL (n = 27) by multi-color flow cytometry and qPCR. ALL LSCs expressed CD19 (B4), CD44 (Pgp-1), CD123 (IL-3RA), and CD184 (CXCR4) in all patients tested. Moreover, in various subgroups of patients, LSCs also displayed CD20 (MS4A1) (10/41 = 24%), CD22 (12/20 = 60%), CD33 (Siglec-3) (20/48 = 42%), CD52 (CAMPATH-1) (17/40 = 43%), IL-1RAP (13/29 = 45%), and/or CD135 (FLT3) (4/20 = 20%). CD25 (IL-2RA) and CD26 (DPPIV) were expressed on LSCs in Ph+ ALL exhibiting BCR/ABL1p210, whereas in Ph+ ALL with BCR/ABL1p190, LSCs variably expressed CD25 but did not express CD26. In Ph− ALL, CD34+/CD38− LSCs expressed IL-1RAP in 6/18 patients (33%), but did not express CD25 or CD26. Normal stem cells stained negative for CD25, CD26 and IL-1RAP, and expressed only low amounts of CD52. In xenotransplantation experiments, CD34+/CD38− and CD34+/CD38+ cells engrafted NSG mice after 12–20 weeks, and targeting with antibodies against CD33 and CD52 resulted in reduced engraftment. Together, LSCs in Ph+ and Ph− ALL display unique marker- and target expression profiles. In Ph+ ALL with BCR/ABL1p210, the LSC-phenotype closely resembles the marker-profile of CD34+/CD38− LSCs in chronic myeloid leukemia, confirming the close biologic relationship of these neoplasms. Targeting of LSCs with specific antibodies or related immunotherapies may facilitate LSC eradication in ALL.
Collapse
Affiliation(s)
- Katharina Blatt
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Ingeborg Menzl
- Department of Biomedical Science, Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210 Vienna, Austria
| | - Gregor Eisenwort
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Sabine Cerny-Reiterer
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Harald Herrmann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Radiotherapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Susanne Herndlhofer
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Gabriele Stefanzl
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Daniela Berger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alexandra Keller
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alexander Hauswirth
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Gregor Hoermann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Michael Willmann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department/Clinic for Companion Animals and Horses, Clinic for Small Animals, Clinical Unit of Internal Medicine, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210 Vienna, Austria
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210 Vienna, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Auenbruggerplatz 2, 8036 Graz, Austria
| | - Wolfgang R Sperr
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Christine Mannhalter
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Junia V Melo
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia; Department of Haematology, Imperial College London, Kensington, London SW7 2AZ, United Kingdom
| | - Ulrich Jäger
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Veronika Sexl
- Department of Biomedical Science, Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210 Vienna, Austria
| | - Peter Valent
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
23
|
Willmann M, Sadovnik I, Eisenwort G, Entner M, Bernthaler T, Stefanzl G, Hadzijusufovic E, Berger D, Herrmann H, Hoermann G, Valent P, Rülicke T. Evaluation of cooperative antileukemic effects of nilotinib and vildagliptin in Ph + chronic myeloid leukemia. Exp Hematol 2017; 57:50-59.e6. [PMID: 29031704 DOI: 10.1016/j.exphem.2017.09.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 09/17/2017] [Accepted: 09/30/2017] [Indexed: 12/13/2022]
Abstract
Chronic myeloid leukemia (CML) is a stem cell (SC) neoplasm characterized by the BCR/ABL1 oncogene. Although the disease can be kept under control using BCR/ABL1 tyrosine kinase inhibitors (TKIs) in most cases, some patients relapse or have resistant disease, so there is a need to identify new therapeutic targets in this malignancy. Recent data suggest that leukemic SCs (LSCs) in CML display the stem-cell (SC)-mobilizing cell surface enzyme dipeptidyl-peptidase IV (DPPIV = CD26) in an aberrant manner. In the present study, we analyzed the effects of the DPPIV blocker vildagliptin as single agent or in combination with the BCR/ABL1 TKI imatinib or nilotinib on growth and survival of CML LSCs in vitro and on LSC engraftment in an in vivo xenotransplantation nonobese diabetic SCID-IL-2Rγ-/- (NSG) mouse model. We found that nilotinib induces apoptosis in CML LSCs and inhibits their engraftment in NSG mice. In contrast, no substantial effects were seen with imatinib or vildagliptin. Nevertheless, vildagliptin was found to reduce the "mobilization" of CML LSCs from a stroma cell layer consisting of mouse fibroblasts in an in vitro co-culture model, suggesting reduced disease expansion. However, although vildagliptin and nilotinib produced cooperative effects in individual experiments, overall, no significant effects of coadministered vildagliptin over nilotinib or imatinib treatment alone were seen on the engraftment of CML cells in NSG mice. Gliptins may be interesting drugs in the context of CML and nilotinib therapy, but our preclinical studies did not reveal a major cooperative effect of the drug-combination vildagliptin + nilotinib on engraftment of CML cells in NSG mice.
Collapse
MESH Headings
- Adamantane/administration & dosage
- Adamantane/analogs & derivatives
- Adamantane/pharmacology
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Apoptosis
- Coculture Techniques
- Dipeptidyl Peptidase 4/drug effects
- Dipeptidyl-Peptidase IV Inhibitors/administration & dosage
- Dipeptidyl-Peptidase IV Inhibitors/pharmacology
- Drug Synergism
- Fibroblasts
- Fusion Proteins, bcr-abl/drug effects
- Humans
- Imatinib Mesylate/pharmacology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Molecular Targeted Therapy
- Neoplasm Proteins/antagonists & inhibitors
- Nitriles/administration & dosage
- Nitriles/pharmacology
- Protein Kinase Inhibitors/administration & dosage
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/administration & dosage
- Pyrimidines/pharmacology
- Pyrrolidines/administration & dosage
- Pyrrolidines/pharmacology
- Tumor Cells, Cultured
- Vildagliptin
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Michael Willmann
- Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria; Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria.
| | - Irina Sadovnik
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Gregor Eisenwort
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria; Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Martin Entner
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tina Bernthaler
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gabriele Stefanzl
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria; Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Emir Hadzijusufovic
- Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria; Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria; Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Daniela Berger
- Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Harald Herrmann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria; Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria; Department of Radiotherapy, Medical University of Vienna, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria; Department of Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Thomas Rülicke
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria; Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
24
|
Hoseini SS, Cheung NK. Acute myeloid leukemia targets for bispecific antibodies. Blood Cancer J 2017; 7:e522. [PMID: 28157217 PMCID: PMC5386336 DOI: 10.1038/bcj.2017.2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022] Open
Abstract
Despite substantial gains in our understanding of the genomics of acute myelogenous leukemia (AML), patient survival remains unsatisfactory especially among the older age group. T cell-based therapy of lymphoblastic leukemia is rapidly advancing; however, its application in AML is still lagging behind. Bispecific antibodies can redirect polyclonal effector cells to engage chosen targets on leukemia blasts. When the effector cells are natural-killer cells, both antibody-dependent and antibody-independent mechanisms could be exploited. When the effectors are T cells, direct tumor cytotoxicity can be engaged followed by a potential vaccination effect. In this review, we summarize the AML-associated tumor targets and the bispecific antibodies that have been studied. The potentials and limitations of each of these systems will be discussed.
Collapse
Affiliation(s)
- S S Hoseini
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - N K Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
25
|
Schulenburg A, Blatt K, Cerny-Reiterer S, Sadovnik I, Herrmann H, Marian B, Grunt TW, Zielinski CC, Valent P. Cancer stem cells in basic science and in translational oncology: can we translate into clinical application? J Hematol Oncol 2015; 8:16. [PMID: 25886184 PMCID: PMC4345016 DOI: 10.1186/s13045-015-0113-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/14/2015] [Indexed: 02/08/2023] Open
Abstract
Since their description and identification in leukemias and solid tumors, cancer stem cells (CSC) have been the subject of intensive research in translational oncology. Indeed, recent advances have led to the identification of CSC markers, CSC targets, and the preclinical and clinical evaluation of the CSC-eradicating (curative) potential of various drugs. However, although diverse CSC markers and targets have been identified, several questions remain, such as the origin and evolution of CSC, mechanisms underlying resistance of CSC against various targeted drugs, and the biochemical basis and function of stroma cell-CSC interactions in the so-called ‘stem cell niche.’ Additional aspects that have to be taken into account when considering CSC elimination as primary treatment-goal are the genomic plasticity and extensive subclone formation of CSC. Notably, various cell fractions with different combinations of molecular aberrations and varying proliferative potential may display CSC function in a given neoplasm, and the related molecular complexity of the genome in CSC subsets is considered to contribute essentially to disease evolution and acquired drug resistance. In the current article, we discuss new developments in the field of CSC research and whether these new concepts can be exploited in clinical practice in the future.
Collapse
Affiliation(s)
- Axel Schulenburg
- Bone Marrow Transplantation Unit, Department of Internal Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, A-1090, Wien, Austria. .,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Stem Cell Transplantation Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Wien, Austria.
| | - Katharina Blatt
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Sabine Cerny-Reiterer
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Irina Sadovnik
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Harald Herrmann
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Radiation Therapy, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria.
| | - Brigitte Marian
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Institute for Cancer Research, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Thomas W Grunt
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Clinical Oncology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Christoph C Zielinski
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Clinical Oncology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| | - Peter Valent
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Spitalgasse 23, Vienna, 1090, Wien, Austria. .,Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Wien, Austria.
| |
Collapse
|
26
|
Valent P, Sadovnik I, Ráčil Z, Herrmann H, Blatt K, Cerny-Reiterer S, Eisenwort G, Lion T, Holyoake T, Mayer J. DPPIV (CD26) as a novel stem cell marker in Ph+ chronic myeloid leukaemia. Eur J Clin Invest 2014; 44:1239-45. [PMID: 25371066 DOI: 10.1111/eci.12368] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/31/2014] [Indexed: 12/21/2022]
Abstract
The concept of leukaemic stem cells (LSCs) has been developed to explain the complex cellular hierarchy and biology of leukaemias and to screen for pivotal targets that can be employed to improve drug therapies through LSC eradication in these patients. Some of the newly discovered LSC markers seem to be expressed in a disease-specific manner and may thus serve as major research tools and diagnostic parameters. A useful LSC marker in chronic myeloid leukaemia (CML) appears to be CD26, also known as dipeptidylpeptidase IV. Expression of CD26 is largely restricted to CD34(+) /CD38(-) LSCs in BCR/ABL1(+) CML, but is not found on LSCs in other myeloid or lymphoid neoplasms, with the exception of lymphoid blast crisis of CML, BCR/ABL1p210 + acute lymphoblastic leukaemia, and a very few cases of acute myeloid leukaemia. Moreover, CD26 usually is not expressed on normal bone marrow (BM) stem cells. Functionally, CD26 is a cytokine-targeting surface enzyme that may facilitate the mobilization of LSCs from the BM niche. In this article, we review our current knowledge about the biology and function of CD26 on CML LSCs and discuss the diagnostic potential of this new LSC marker in clinical haematology.
Collapse
Affiliation(s)
- Peter Valent
- Division of Haematology & Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|