1
|
Kegyes D, Thiagarajan PS, Ghiaur G. MRD in Acute Leukemias: Lessons Learned from Acute Promyelocytic Leukemia. Cancers (Basel) 2024; 16:3208. [PMID: 39335179 PMCID: PMC11430625 DOI: 10.3390/cancers16183208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Introduction: Advances in molecular biology, polymerase chain reaction (PCR), and next-generation sequencing (NGS) have transformed the concept of minimal residual disease (MRD) from a philosophical idea into a measurable reality. Current Treatment Paradigms and Lessons Learned from APL: Acute promyelocytic leukemia (APL) leads the way in this transformation, initially using PCR to detect MRD in patients in remission, and more recently, aiming to eliminate it entirely with modern treatment strategies. Along the way, we have gained valuable insights that, when applied to other forms of acute leukemia, hold the potential to significantly improve the outcomes of these challenging diseases. Does the BM Microenvironment Play a Role in MRD?: In this review, we explore the current use of MRD in the management of acute leukemia and delve into the biological processes that contribute to MRD persistence, including its overlap with leukemia stem cells and the role of the bone marrow microenvironment.
Collapse
Affiliation(s)
- David Kegyes
- MedFuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
- The Sidney Kimmel Cancer Center, The Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Gabriel Ghiaur
- The Sidney Kimmel Cancer Center, The Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
2
|
Karantanos T, Teodorescu P, Perkins B, Christodoulou I, Esteb C, Varadhan R, Helmenstine E, Rajkhowa T, Paun BC, Bonifant C, Dalton WB, Gondek LP, Moliterno AR, Levis MJ, Ghiaur G, Jones RJ. The role of the atypical chemokine receptor CCRL2 in myelodysplastic syndrome and secondary acute myeloid leukemia. SCIENCE ADVANCES 2022; 8:eabl8952. [PMID: 35179961 PMCID: PMC8856621 DOI: 10.1126/sciadv.abl8952] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/23/2021] [Indexed: 06/06/2023]
Abstract
The identification of new pathways supporting the myelodysplastic syndrome (MDS) primitive cells growth is required to develop targeted therapies. Within myeloid malignancies, men have worse outcomes than women, suggesting male sex hormone-driven effects in malignant hematopoiesis. Androgen receptor promotes the expression of five granulocyte colony-stimulating factor receptor-regulated genes. Among them, CCRL2 encodes an atypical chemokine receptor regulating cytokine signaling in granulocytes, but its role in myeloid malignancies is unknown. Our study revealed that CCRL2 is up-regulated in primitive cells from patients with MDS and secondary acute myeloid leukemia (sAML). CCRL2 knockdown suppressed MDS92 and MDS-L cell growth and clonogenicity in vitro and in vivo and decreased JAK2/STAT3/STAT5 phosphorylation. CCRL2 coprecipitated with JAK2 and potentiated JAK2-STAT interaction. Erythroleukemia cells expressing JAK2V617F showed less effect of CCRL2 knockdown, whereas fedratinib potentiated the CCRL2 knockdown effect. Conclusively, our results implicate CCRL2 as an MDS/sAML cell growth mediator, partially through JAK2/STAT signaling.
Collapse
Affiliation(s)
- Theodoros Karantanos
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Patric Teodorescu
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Brandy Perkins
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Ilias Christodoulou
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Christopher Esteb
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics, Johns Hopkins/Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Eric Helmenstine
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Trivikram Rajkhowa
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Bogdan C. Paun
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Challice Bonifant
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - W. Brian Dalton
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Lukasz P. Gondek
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Alison R. Moliterno
- Division of Adult Hematology, Department of Medicine, Johns Hopkins University, Baltimore MD, USA
| | - Mark J. Levis
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Gabriel Ghiaur
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Richard J. Jones
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, USA
| |
Collapse
|
3
|
Functional genomic approaches in acute myeloid leukemia: Insights into disease models and the therapeutic potential of reprogramming. Cancer Lett 2022; 533:215579. [DOI: 10.1016/j.canlet.2022.215579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/17/2022] [Accepted: 01/29/2022] [Indexed: 11/19/2022]
|
4
|
Touil Y, Latreche-Carton C, Bouazzati HE, Nugues AL, Jouy N, Thuru X, Laine W, Lepretre F, Figeac M, Tardivel M, Kluza J, Idziorek T, Quesnel B. p65/RelA NF-κB fragments generated by RIPK3 activity regulate tumorigenicity, cell metabolism, and stemness characteristics. J Cell Biochem 2021; 123:543-556. [PMID: 34927768 PMCID: PMC9299825 DOI: 10.1002/jcb.30198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/17/2021] [Accepted: 12/10/2021] [Indexed: 12/16/2022]
Abstract
Receptor‐interacting protein kinase 3 (RIPK3) can induce necroptosis, apoptosis, or cell proliferation and is silenced in several hematological malignancies. We previously reported that RIPK3 activity independent of its kinase domain induces caspase‐mediated p65/RelA cleavage, resulting in N‐terminal 1‐361 and C‐terminal 362‐549 fragments. We show here that a noncleavable p65/RelA D361E mutant expressed in DA1‐3b leukemia cells decreases mouse survival times and that coexpression of p65/RelA fragments increases the tumorigenicity of B16F1 melanoma cells. This aggressiveness in vivo did not correlate with NF‐κB activity measured in vitro. The fragments and p65/RelA D361E mutant induced different expression profiles in DA1‐3b and B16F1 cells. Stemness markers were affected: p65/RelA D361E increased ALDH activity in DA1‐3b cells, and fragment expression increased melanoma sphere formation in B16/F1 cells. p65/RelA fragments and the D361E noncleavable mutant decreased oxidative or glycolytic cell metabolism, with differences observed between models. Thus, p65/RelA cleavage initiated by kinase‐independent RIPK3 activity in cancer cells is not neutral and induces pleiotropic effects in vitro and in vivo that may vary across tumor types.
Collapse
Affiliation(s)
- Yasmine Touil
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Céline Latreche-Carton
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Hassiba El Bouazzati
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Anne-Lucie Nugues
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Nathalie Jouy
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Xavier Thuru
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - William Laine
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | | | - Martin Figeac
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Meryem Tardivel
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Jérôme Kluza
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Thierry Idziorek
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Bruno Quesnel
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France.,Service des Maladies du Sang, CHU Lille, Lille, France
| |
Collapse
|
5
|
Naz F, Shi M, Sajid S, Yang Z, Yu C. Cancer stem cells: a major culprit of intra-tumor heterogeneity. Am J Cancer Res 2021; 11:5782-5811. [PMID: 35018226 PMCID: PMC8727794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/25/2021] [Indexed: 06/14/2023] Open
Abstract
Cancer is recognized as a preeminent factor of the world's mortality. Although various modalities have been designed to cure this life-threatening ailment, a significant impediment in the effective output of cancer treatment is heterogeneity. Cancer is characterized as a heterogeneous health disorder that comprises a distinct group of transformed cells to assist anomalous proliferation of affected cells. Cancer stem cells (CSCs) are a leading cause of cancer heterogeneity that is continually transformed by cellular extrinsic and intrinsic factors. They intensify neoplastic cells aggressiveness by strengthening their dissemination, relapse and therapy resistance. Considering this viewpoint, in this review article we have discussed some intrinsic (transcription factors, cell signaling pathways, genetic alterations, epigenetic modifications, non-coding RNAs (ncRNAs) and epitranscriptomics) and extrinsic factors (tumor microenvironment (TME)) that contribute to CSC heterogeneity and plasticity, which may help scientists to meddle these processes and eventually improve cancer research and management. Besides, the potential role of CSCs heterogeneity in establishing metastasis and therapy resistance has been articulated which signifies the importance of developing novel anticancer therapies to target CSCs along with targeting bulk tumor mass to achieve an effective output.
Collapse
Affiliation(s)
- Faiza Naz
- College of Life Science and Technology, Beijing University of Chemical TechnologyBeijing 100029, China
| | - Mengran Shi
- College of Life Science and Technology, Beijing University of Chemical TechnologyBeijing 100029, China
| | - Salvia Sajid
- Department of Biotechnology, Jinnah University for WomenKarachi 74600, Pakistan
| | - Zhao Yang
- College of Life Science and Technology, Beijing University of Chemical TechnologyBeijing 100029, China
- College of Life Science, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim UniversityAlar 843300, Xinjiang, China
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical TechnologyBeijing 100029, China
| |
Collapse
|
6
|
Webster JA, Yogarajah M, Zahurak M, Symons H, Dezern AE, Gojo I, Prince GT, Morrow J, Jones RJ, Smith BD, Showel M. A phase II study of azacitidine in combination with granulocyte-macrophage colony-stimulating factor as maintenance treatment, after allogeneic blood or marrow transplantation in patients with poor-risk acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Leuk Lymphoma 2021; 62:3181-3191. [PMID: 34284701 PMCID: PMC9195564 DOI: 10.1080/10428194.2021.1948029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/14/2021] [Accepted: 06/17/2021] [Indexed: 10/20/2022]
Abstract
Relapse is the most common cause of treatment failure following allogeneic blood or marrow transplantation (alloBMT) for AML or MDS. Post-transplant maintenance therapies may prevent relapse. We conducted a phase II trial combining azacitidine (AZA) with GM-CSF in non-relapsed, post-transplant patients with AML or MDS. Patients received escalating doses of AZA to a maximum of 75 mg/m2 for 5 days per cycle for up to 12 cycles. GM-CSF was given on days 1-10 of each cycle. Eighteen patients were treated following non-myeloablative (17) and myeloablative (1) alloBMT for AML (61.1%), MDS (27.7%), or therapy-related myeloid neoplasm (11.1%). The majority of patients (72%) received their graft from an HLA-haploidentical donor. The treatment was well-tolerated with rare grade 3-4 hematologic toxicities. One patient suffered an exacerbation of GVHD. The 24-month relapse-free and overall survivals were 47 and 57%, respectively, with a median of 18.6 and 29 months.
Collapse
Affiliation(s)
- Jonathan A Webster
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Marianna Zahurak
- Division of Biostatistics and Bioinformatics, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Heather Symons
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy E Dezern
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ivana Gojo
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gabrielle T Prince
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Morrow
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard J Jones
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B Douglas Smith
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Margaret Showel
- Hematologic Malignancies and Bone Marrow Transplantation Program, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Huang Q, Zhong J, Gao H, Li K, Liang H. Subgrouping by gene expression profiles to improve relapse risk prediction in paediatric B-precursor acute lymphoblastic leukaemia. Cancer Med 2021; 10:3782-3793. [PMID: 33987975 PMCID: PMC8178509 DOI: 10.1002/cam4.3842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 11/17/2020] [Accepted: 02/22/2021] [Indexed: 11/08/2022] Open
Abstract
Relapsed acute lymphoblastic leukaemia (ALL) remains a prevalent paediatric cancer and one of the most common causes of mortality from malignancy in children. Tailoring the intensity of therapy according to early stratification is a promising strategy but remains a major challenge due to heterogeneity and subtyping difficulty. In this study, we subgroup B-precursor ALL patients by gene expression profiles, using non-negative matrix factorization and minimum description length which unsupervisedly determines the number of subgroups. Within each of the four subgroups, logistic and Cox regression with elastic net regularization are used to build models predicting minimal residual disease (MRD) and relapse-free survival (RFS) respectively. Measured by area under the receiver operating characteristic curve (AUC), subgrouping improves prediction of MRD in one subgroup which mostly overlaps with subtype TCF3-PBX1 (AUC = 0·986 in the training set and 1·0 in the test set), compared to a global model published previously. The models predicting RFS displayed acceptable concordance in training set and discriminate high-relapse-risk patients in three subgroups of the test set (Wilcoxon test p = 0·048, 0·036, and 0·016). Genes playing roles in the models are specific to different subgroups. The improvement of subgrouped MRD prediction and the differences of genes in prediction models of subgroups suggest that the heterogeneity of B-precursor ALL can be handled by subgrouping according to gene expression profiles to improve the prediction accuracy.
Collapse
Affiliation(s)
- Qingsheng Huang
- School of Mathematics and Statistics, Hanshan Normal University, Chaozhou, China.,Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Jiayong Zhong
- Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Huan Gao
- Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Kuanrong Li
- Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Huiying Liang
- Clinical Data Center, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
8
|
Yanagisawa B, Perkins B, Karantanos T, Levis M, Ghiaur G, Smith BD, Jones RJ. Expression of putative leukemia stem cell targets in genetically-defined acute myeloid leukemia subtypes. Leuk Res 2020; 99:106477. [PMID: 33220589 DOI: 10.1016/j.leukres.2020.106477] [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: 09/02/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 01/07/2023]
Abstract
Although most acute myeloid leukemia (AML) patients achieve complete remissions, the majority still eventually relapse and die of their disease. Rare primitive leukemia cells, so-called leukemia stem cells (LSCs), represent one potential type of resistant cell subpopulation responsible for this dissociation between response and cure. Several LSC targets have been described, but there is limited evidence about their relative utility or that targeting any can prevent relapse. LSCs not only appear to be biologically heterogeneous, but the classic immunocompromised mouse transplantation model also has serious shortcomings as an LSC assay. Out data suggest that the most immature cell phenotype that can be identified within a patient's leukemia may be clinically relevant and represent the de facto LSC. Moreover, although phenotypically heterogeneous, these putative LSCs show consistent phenotypes within individual genetically defined groups. Using this LSC definition, we studied several previously described putative LSC targets, CD25, CD26, CD47, CD96, CD123, and CLL-1, and all were expressed across heterogeneous LSC phenotypes. In addition, with the exception of CD47, there was at most low expression of these targets on normal hematopoietic stem cells (HSCs). CD123 and CLL-1 demonstrated the greatest expression differences between putative LSCs and normal HSCs. Importantly, CD123 monoclonal antibodies were cytotoxic in vitro to putative LSCs from all AML subtypes, while showing limited to no toxicity against normal HSCs and hematopoietic progenitors. Since minimal residual disease appears to be a more homogeneous population of cells responsible for relapse, targeting CD123 in this setting may be most effective.
Collapse
Affiliation(s)
- Breann Yanagisawa
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Brandy Perkins
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | - Mark Levis
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Gabriel Ghiaur
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - B Douglas Smith
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Richard J Jones
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| |
Collapse
|
9
|
Yang L, Chen WM, Dao FT, Zhang YH, Wang YZ, Chang Y, Liu YR, Jiang Q, Zhang XH, Liu KY, Huang XJ, Qin YZ. High aldehyde dehydrogenase activity at diagnosis predicts relapse in patients with t(8;21) acute myeloid leukemia. Cancer Med 2019; 8:5459-5467. [PMID: 31364309 PMCID: PMC6745853 DOI: 10.1002/cam4.2422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/19/2019] [Accepted: 07/02/2019] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukemia (AML) with t(8;21) is a heterogeneous disease. Although the detection of minimal residual disease (MRD), which is indicated by RUNX1‐RUNX1T1 transcript levels, plays a key role in directing treatment, risk stratification needs to be improved, and other markers need to be assessed. A total of 66 t(8;21) AML patients were tested for aldehyde dehydrogenase (ALDH) activity by flow cytometry at diagnosis, and 52 patients were followed up for a median of 20 (1‐34) months. The median percentage of CD34+ALDH+, CD34+CD38‐ALDH+, and CD34+CD38+ALDH+ cells among nucleated cells were 0.028%, 0.012%, and 0.0070%, respectively. The CD34+ALDH+‐H, CD34+CD38‐ALDH+‐H, and CD34+CD38+ALDH+‐H statuses (the percentage of cells that were higher than the individual cutoffs) were all significantly associated with a lower 2‐year relapse‐free survival (RFS) rate in both the whole cohort and adult patients (P = .015, .016, and .049; P = .014, .018, and .032). Patients with < 3‐log reduction in the RUNX1‐RUNX1T1 transcript level after the second consolidation therapy (defined as MRD‐H) had a significantly lower 2‐year RFS rate than patients with ≥ 3‐log reduction (MRD‐L) (P = .017). The CD34+ALDH+ status at diagnosis was then combined with the MRD status. CD34+ALDH+‐L/MRD‐H patients had similar 2‐year RFS rates to both CD34+ALDH+‐L/MRD‐L and CD34+ALDH+‐H/MRD‐L patients (P = .50 and 1.0); and CD34+ALDH+‐H/MRD‐H patients had significantly lower 2‐year RFS rate compared with CD34+ALDH+‐L and/or MRD‐L patients (P < .0001). Multivariate analysis showed that CD34+ALDH+‐H/MRD‐H was an independent adverse prognostic factor for relapse. In conclusion, ALDH status at diagnosis may improve MRD‐based risk stratification in t(8;21) AML, and concurrent high levels of CD34+ALDH+ at diagnosis and MRD predict relapse.
Collapse
Affiliation(s)
- Lu Yang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Wen-Min Chen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Feng-Ting Dao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Yan-Huan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Ya-Zhe Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Yan Chang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Yan-Rong Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qian Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Ya-Zhen Qin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| |
Collapse
|
10
|
Acute Myeloid Leukemia Stem Cell Heterogeneity and Its Clinical Relevance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1139:153-169. [DOI: 10.1007/978-3-030-14366-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
11
|
Moshaver B, Wouters RF, Kelder A, Ossenkoppele GJ, Westra GAH, Kwidama Z, Rutten AR, Kaspers GJL, Zweegman S, Cloos J, Schuurhuis GJ. Relationship between CD34/CD38 and side population (SP) defined leukemia stem cell compartments in acute myeloid leukemia. Leuk Res 2019; 81:27-34. [PMID: 31002948 DOI: 10.1016/j.leukres.2019.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 10/27/2022]
Abstract
Leukemic stem cells (LSCs), defined by CD34/CD38 expression, are believed to be essential for leukemia initiation and therapy resistance in acute myeloid leukemia. In addition, the side population (SP), characterized by high Hoechst 33342 efflux, reflecting therapy resistance, has leukemia initiating ability. The purpose of this study is, in both CD34-positive and CD34-negative AML, to integrate both types of LSC compartment into a new more restricted definition. Different CD34/CD38/SP defined putative LSC and normal hematopoietic compartments, with neoplastic or normal nature, respectively, were thus identified after cell sorting, and confirmed by FISH/PCR. Stem cell activity was assessed in the long-term liquid culture stem cell assay. SP fractions harbored the strongest functional stem cell activity in both normal and neoplastic cells in both CD34-positive and CD34-negative AML. Overall, inclusion of SP fraction decreased the size of the putative CD34/CD38 defined LSC compartment by a factor >500. For example, for the important CD34+CD38- LSC compartment, the median SP/CD34+CD38- frequency was 5.1 per million WBC (CD34-positive AML), and median SP/CD34-CD38+ frequency (CD34-negative AML) was 1796 per million WBC. Improved detection of LSC may enable identification of therapy resistant clones, and thereby identification of novel LSC specific, HSC sparing, therapies.
Collapse
Affiliation(s)
- Bijan Moshaver
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Rolf F Wouters
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands; Department of Pediatric Oncology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Angèle Kelder
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Gert J Ossenkoppele
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Guus A H Westra
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Zinia Kwidama
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands; Department of Pediatric Oncology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Arjo R Rutten
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Gert J L Kaspers
- Department of Pediatric Oncology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Jacqueline Cloos
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands; Department of Pediatric Oncology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands
| | - Gerrit J Schuurhuis
- Department of Hematology, Cancer Center Amsterdam, VU University Medical Center, 1081HV Amsterdam, the Netherlands.
| |
Collapse
|
12
|
Blume R, Rempel E, Manta L, Saeed BR, Wang W, Raffel S, Ermakova O, Eckstein V, Benes V, Trumpp A, Ho AD, Lutz C. The molecular signature of AML with increased ALDH activity suggests a stem cell origin. Leuk Lymphoma 2018; 59:2201-2210. [DOI: 10.1080/10428194.2017.1422862] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Rachel Blume
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Eugen Rempel
- Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Linda Manta
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Borhan R. Saeed
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Wenwen Wang
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Simon Raffel
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Olga Ermakova
- Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Volker Eckstein
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Vladimir Benes
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Anthony D. Ho
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| | - Christoph Lutz
- Department of Medicine V, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
13
|
ALDHs in normal and malignant hematopoietic cells: Potential new avenues for treatment of AML and other blood cancers. Chem Biol Interact 2017. [PMID: 28645468 DOI: 10.1016/j.cbi.2017.06.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multiple studies have demonstrated that ALDH1A1 is elevated in hematopoietic stem cells (HSCs). As a means to better characterize such cells, we previously developed the fluorescent ALDH1A1 substrate Aldefluor to facilitate HSC identification and isolation. This has proven useful for counting and isolating HSCs from human bone marrow, peripheral blood and cord blood as well as stem cells in other tissues and organisms. Given the high level expression of ALDH1A1, we explored its biology and that of other ALDHs in HSCs and found that ALDH1A1 and ALDH3A1 were important in metabolizing reactive aldehydes (RAlds) and reactive oxygen species (ROS). In murine models, loss of these two isoforms resulted in a variety of effects on HSC biology, increased DNA damage and predisposition to leukemia formation when combined with a genetic driver of HSC proliferation and self-renewal. Loss of ALDH activity may also predispose to marrow failure and AML in Fanconi's anemia (FA). ALDHs also have importance in mediating drug resistance in AML, may be useful in the identification of leukemia stem cells (LSCs) and ALDH activity levels may have prognostic significance. Together these findings suggest that further studying ALDH biology in AML and other blood cancers may provide important insights into malignant transformation and may point the way to the development of novel diagnostics and therapies.
Collapse
|
14
|
Jentzsch M, Bill M, Nicolet D, Leiblein S, Schubert K, Pless M, Bergmann U, Wildenberger K, Schuhmann L, Cross M, Pönisch W, Franke GN, Vucinic V, Lange T, Behre G, Mrózek K, Bloomfield CD, Niederwieser D, Schwind S. Prognostic impact of the CD34+/CD38- cell burden in patients with acute myeloid leukemia receiving allogeneic stem cell transplantation. Am J Hematol 2017; 92:388-396. [PMID: 28133783 DOI: 10.1002/ajh.24663] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 01/09/2023]
Abstract
In acute myeloid leukemia (AML), leukemia-initiating cells exist within the CD34+/CD38- cell compartment. They are assumed to be more resistant to chemotherapy, enriched in minimal residual disease cell populations, and responsible for relapse. Here we evaluated clinical and biological associations and the prognostic impact of a high diagnostic CD34+/CD38- cell burden in 169 AML patients receiving an allogeneic stem cell transplantation in complete remission. Here, the therapeutic approach is mainly based on immunological graft-versus-leukemia effects. Percentage of bone marrow CD34+/CD38- cell burden at diagnosis was measured using flow cytometry and was highly variable (median 0.5%, range 0%-89% of all mononuclear cells). A high CD34+/CD38- cell burden at diagnosis associated with worse genetic risk and secondary AML. Patients with a high CD34+/CD38- cell burden had shorter relapse-free and overall survival which may be mediated by residual leukemia-initiating cells in the CD34+/CD38- cell population, escaping the graft-versus-leukemia effect after allogeneic transplantation. Evaluating the CD34+/CD38- cell burden at diagnosis may help to identify patients at high risk of relapse after allogeneic transplantation. Further studies to understand leukemia-initiating cell biology and develop targeting therapies to improve outcomes of AML patients are needed.
Collapse
Affiliation(s)
- Madlen Jentzsch
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Marius Bill
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center; Columbus Ohio USA
| | - Sabine Leiblein
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Karoline Schubert
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Martina Pless
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Ulrike Bergmann
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Kathrin Wildenberger
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Luba Schuhmann
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Michael Cross
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Wolfram Pönisch
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Georg-Nikolaus Franke
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Vladan Vucinic
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Thoralf Lange
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Gerhard Behre
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center; Columbus Ohio USA
| | | | - Dietger Niederwieser
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| | - Sebastian Schwind
- Department of Hematology; Oncology and Hemostaseology, University of Leipzig; Leipzig Germany
| |
Collapse
|
15
|
Yanagisawa B, Ghiaur G, Smith BD, Jones RJ. Translating leukemia stem cells into the clinical setting: Harmonizing the heterogeneity. Exp Hematol 2016; 44:1130-1137. [PMID: 27693555 PMCID: PMC5110366 DOI: 10.1016/j.exphem.2016.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/23/2016] [Indexed: 01/01/2023]
Abstract
Considerable evidence suggests that rare leukemia cells with stem cell features, including self-renewal capacity and drug resistance, are primarily responsible for both disease maintenance and relapses. Traditionally, these so-called leukemia stem cells (LSCs) have been identified in the laboratory by their ability to engraft acute myeloid leukemia (AML) into immunocompromised mice. For many years, only those rare AML cells characterized by a hematopoietic stem cell (HSC) CD34+CD38- phenotype were believed capable of generating leukemia in immunocompromised mice. However, more recently, significant heterogeneity in the phenotypes of those AML cells that can engraft immunocompromised mice has been demonstrated. AML cells that engraft immunocompromised mice have also been shown to not necessarily represent either the founder clone or those cells responsible for relapse. A recent study found that the most immature phenotype present in an AML correlated with genetically defined risk groups and outcomes, but was heterogeneous. Patients with AML cells expressing a primitive HSC phenotype (CD34+CD38- with high aldehyde dehydrogenase activity) manifested significantly lower complete remission rates, as well as poorer event-free and overall survivals. Leukemias in which the most primitive cells displayed more mature phenotypes were associated with better outcomes. The strong clinical correlations suggest that the most immature phenotype detectable within a patient's AML might serve as a biomarker for "clinically relevant" LSCs.
Collapse
Affiliation(s)
- Breann Yanagisawa
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Gabriel Ghiaur
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - B Douglas Smith
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Richard J Jones
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|