1
|
Li H, Cao Z, Liu Y, Xue Z, Li Y, Xing H, Xu Y, Gu R, Qiu S, Wei H, Wang M, Rao Q, Wang J. Slow-replicating leukemia cells represent a leukemia stem cell population with high cell-surface CD74 expression. Mol Oncol 2024; 18:2554-2568. [PMID: 38922758 PMCID: PMC11459046 DOI: 10.1002/1878-0261.13690] [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: 12/07/2023] [Revised: 04/25/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Persistence of quiescent leukemia stem cells (LSCs) after treatment most likely contributes to chemotherapy resistance and poor prognosis of leukemia patients. Identification of this quiescent cell population would facilitate eradicating LSCs. Here, using a cell-tracing PKH26 (PKH) dye that can be equally distributed to daughter cells following cell division in vivo, we identify a label-retaining slow-cycling leukemia cell population from AML1-ETO9a (AE9a) leukemic mice. We find that, compared with cells not maintaining PKH-staining, a higher proportion of PKH-retaining cells are in G0 phase, and PKH-retaining cells exhibit increased colony formation ability and leukemia initiation potential. In addition, PKH-retaining cells possess high chemo-resistance and are more likely to be localized to the endosteal bone marrow region. Based on the transcriptional signature, HLA class II histocompatibility antigen gamma chain (Cd74) is highly expressed in PKH-retaining leukemia cells. Furthermore, cell surface CD74 was identified to be highly expressed in LSCs of AE9a mice and CD34+ human leukemia cells. Compared to Lin-CD74- leukemia cells, Lin-CD74+ leukemia cells of AE9a mice exhibit higher stemness properties. Collectively, our findings reveal that the identified slow-cycling leukemia cell population represents an LSC population, and CD74+ leukemia cells possess stemness properties, suggesting that CD74 is a candidate LSC surface marker.
Collapse
Affiliation(s)
- Huan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Zhijie Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Yiming Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Zhenya Xue
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Yishuang Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Runxia Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Shaowei Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Hui Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| |
Collapse
|
2
|
Zhang Y, Chen L, Fu T, Xu A, Li K, Hao K, Lyu J, Wang Z, Kong F. Self-Stimulated Photodynamic Nanoreactor in Combination with CXCR4 Antagonists for Antileukemia Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21610-21622. [PMID: 38647446 DOI: 10.1021/acsami.4c01603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The treatment of acute myeloid leukemia (AML) remains unsatisfactory, owing to the absence of efficacious therapy regimens over decades. However, advances in molecular biology, including inhibiting the CXCR4/CXCL12 biological axis, have introduced novel therapeutic options for AML. Additionally, self-stimulated phototherapy can solve the poor light penetration from external sources, and it will overcome the limitation that traditional phototherapy cannot be applied to the treatment of AML. Herein, we designed and manufactured a self-stimulated photodynamic nanoreactor to enhance antileukemia efficacy and suppress leukemia recurrence and metastasis in AML mouse models. To fulfill our design, we utilized the CXCR4/CXCL12 biological axis and biomimetic cell membranes in conjunction with self-stimulated phototherapy. This nanoreactor possesses the capability to migrate into the bone marrow cavity, inhibit AML cells from infiltrating into the visceral organ, significantly enhance the antileukemia effect, and prolong the survival time of leukemic mice. Therefore, this nanoreactor has significant potential for achieving high success rates and low recurrence rates in leukemia treatment.
Collapse
MESH Headings
- Animals
- Receptors, CXCR4/metabolism
- Receptors, CXCR4/antagonists & inhibitors
- Mice
- Photochemotherapy
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Photosensitizing Agents/chemistry
- Photosensitizing Agents/pharmacology
- Photosensitizing Agents/therapeutic use
- Cell Line, Tumor
- Chemokine CXCL12/metabolism
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Yan Zhang
- School of Laboratory Medicine, Hangzhou Medical College, 310053 Hangzhou, Zhejiang, China
| | - Liang Chen
- School of Laboratory Medicine, Hangzhou Medical College, 310053 Hangzhou, Zhejiang, China
| | - Ting Fu
- School of Laboratory Medicine, Hangzhou Medical College, 310053 Hangzhou, Zhejiang, China
| | - Aibo Xu
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital, Affiliated People's hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Kaiqiang Li
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital, Affiliated People's hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Ke Hao
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital, Affiliated People's hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Jianxin Lyu
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital, Affiliated People's hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital, Affiliated People's hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Fei Kong
- School of Laboratory Medicine, Hangzhou Medical College, 310053 Hangzhou, Zhejiang, China
- Center for Laboratory Medicine, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital, Affiliated People's hospital, Hangzhou Medical College, Hangzhou 310014, China
| |
Collapse
|
3
|
Bai Y, Sun X, Li M, Niu X, Cao W, Niu J, Xiao X, Chen Y, Sun K. CD7-positive leukemic blasts with DNMT3A mutations predict poor prognosis in patients with acute myeloid leukemia. Front Oncol 2024; 14:1342998. [PMID: 38577341 PMCID: PMC10991683 DOI: 10.3389/fonc.2024.1342998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/15/2024] [Indexed: 04/06/2024] Open
Abstract
Background DNMT3A mutations can be detected in premalignant hematopoietic stem cells and are primarily associated with clonal hematopoiesis of indeterminate potential; however, current evidence does not support assigning them to a distinct European Leukemia Net (ELN) prognostic risk stratification. CD7 is a lymphoid antigen expressed on blasts in approximately 30% of acute myeloid leukemia (AML), and its role in AML remains unclear and depends on subgroup evaluation. This study investigated the prognostic value of DNMT3A mutation combined with CD7 expression in AML. Methods We retrospectively analyzed the clinical data of 297 newly diagnosed non-M3 AML patients. According to the DNMT3A mutation and CD7 expression in AML cells, patients were divided into the DNMT3A-mutated/CD7-positive (CD7+), DNMT3A-mutated/CD7-negative (CD7-), DNMT3A-wild-type/CD7+, and DNMT3A-wild-type/CD7- groups. Results The DNMT3A-mutated/CD7+ group had lower complete remission rates and higher relapse rates. Importantly, these patients had significantly shorter overall survival (OS) and relapse-free survival (RFS). Furthermore, multivariate analysis showed that CD7+ with DNMT3A mutation was an independent risk factor for OS and RFS. Conclusion CD7+ with DNMT3A mutation predicts a poor prognosis in AML patients, and the immunophenotype combined with molecular genetic markers can help to further refine the current risk stratification of AML.
Collapse
Affiliation(s)
- Yanliang Bai
- Department of Hematology, Zhengzhou University People’s Hospital and Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiaobai Sun
- Department of Hematology, Zhengzhou University People’s Hospital and Henan Provincial People’s Hospital, Zhengzhou, China
| | - Mengyi Li
- Department of Hematology, Nanyang Second General Hospital, Nanyang, China
| | - Xiaona Niu
- Department of Hematology, Zhengzhou University People’s Hospital and Henan Provincial People’s Hospital, Zhengzhou, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junwei Niu
- Department of Hematology, Zhengzhou University People’s Hospital and Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xingjun Xiao
- Department of Hematology, Zhengzhou University People’s Hospital and Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yuqing Chen
- Department of Hematology, Zhengzhou University People’s Hospital and Henan Provincial People’s Hospital, Zhengzhou, China
| | - Kai Sun
- Department of Hematology, Zhengzhou University People’s Hospital and Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Hematology, Beijing JiShuiTan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Fan W, Li X. The SIRT1-c-Myc axis in regulation of stem cells. Front Cell Dev Biol 2023; 11:1236968. [PMID: 37554307 PMCID: PMC10405831 DOI: 10.3389/fcell.2023.1236968] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/10/2023] [Indexed: 08/10/2023] Open
Abstract
SIRT1 is the most conserved mammalian NAD+-dependent protein deacetylase. Through deacetylation of transcriptional factors and co-factors, this protein modification enzyme is critically involved in metabolic and epigenetic regulation of stem cells, which is functionally important in maintaining their pluripotency and regulating their differentiation. C-Myc, a key member of Myc proton-oncogene family, is a pivotal factor for transcriptional regulation of genes that control acquisition and maintenance of stemness. Previous cancer research has revealed an intriguing positive feedback loop between SIRT1 and c-Myc that is crucial in tumorigenesis. Recent literature has uncovered important functions of this axis in regulation of maintenance and differentiation of stem cells, including pluripotent stem cells and cancer stem cells. This review highlights recent advances of the SIRT1-c-Myc axis in stem cells.
Collapse
Affiliation(s)
- Wei Fan
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, United States
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, United States
| |
Collapse
|
5
|
Wu C, Cui J, Huo Y, Shi L, Wang C. Alternative splicing of HOXB-AS3 underlie the promoting effect of nuclear m6A reader YTHDC1 on the self-renewal of leukemic stem cells in acute myeloid leukemia. Int J Biol Macromol 2023; 237:123990. [PMID: 36906205 DOI: 10.1016/j.ijbiomac.2023.123990] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/21/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023]
Abstract
This research sought to elucidate the mechanism underlying the self-renewal capacity of leukemic stem cells (LSCs) to offer new insights into the treatment of acute myeloid leukemia (AML). The expression of HOXB-AS3 and YTHDC1 in the AML samples was screened and verified in THP-1 cells and LSCs. The relationship between HOXB-AS3 and YTHDC1 was determined. HOXB-AS3 and YTHDC1 were knocked down through cell transduction to examine the effect of HOXB-AS3 and YTHDC1 on LSCs isolated from THP-1 cells. Tumor formation in mice was used to verify fore experiments. HOXB-AS3 and YTHDC1 were robustly induced in AML, in correlation with adverse prognosis in patients with AML. We found YTHDC1 bound HOXB-AS3 and regulated its expression. Overexpression of YTHDC1 or HOXB-AS3 promoted the proliferation of THP-1 cells and LSCs and impaired their apoptosis, increasing the number of LSCs in the blood and bone marrow of AML mice. YTHDC1 could upregulate the expression of HOXB-AS3 spliceosome NR_033205.1 via the m6A modification of HOXB-AS3 precursor RNA. By this mechanism, YTHDC1 accelerated the self-renewal of LSCs and the subsequent AML progression. This study identifies a crucial role for YTHDC1 in the regulation of LSC self-renewal in AML and suggests a new perspective for AML treatment.
Collapse
Affiliation(s)
- Chuan Wu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jieke Cui
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yankun Huo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Luyao Shi
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Chong Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
6
|
Lee JC, Liu S, Wang Y, Liang Y, Jablons DM. MK256 is a novel CDK8 inhibitor with potent antitumor activity in AML through downregulation of the STAT pathway. Oncotarget 2022; 13:1217-1236. [PMCID: PMC9629815 DOI: 10.18632/oncotarget.28305] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most lethal form of AML due to disease relapse. Cyclin dependent kinase 8 (CDK8) is a serine/threonine kinase that belongs to the family of Cyclin-dependent kinases and is an emerging target for the treatment of AML. MK256, a potent, selective, and orally available CDK8 inhibitor was developed to target AML. We sought to examine the anticancer effect of MK256 on AML. In CD34+/CD38- leukemia stem cells, we found that MK256 induced differentiation and maturation. Treatment of MK256 inhibited proliferation of AML cell lines. Further studies of the inhibitory effect suggested that MK256 not only downregulated phosphorylated STAT1(S727) and STAT5(S726), but also lowered mRNA expressions of MCL-1 and CCL2 in AML cell lines. Efficacy of MK256 was shown in MOLM-14 xenograft models, and the inhibitory effect on phosphorylated STAT1(S727) and STAT5(S726) with treatment of MK256 was observed in vivo. Pharmacologic dynamics study of MK256 in MOLM-14 xenograft models showed dose-dependent inhibition of the STAT pathway. Both in vitro and in vivo studies suggested that MK256 could effectively downregulate the STAT pathway. In vitro ADME, pharmacological kinetics, and toxicity of MK256 were profiled to evaluate the drug properties of MK256. Our results show that MK256 is a novel CDK8 inhibitor with a desirable efficacy and safety profile and has great potential to be a promising drug candidate for AML through regulating the STAT pathway.
Collapse
Affiliation(s)
- Jen-Chieh Lee
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA,2Touro University, College of Osteopathic Medicine, Vallejo, CA 94592, USA,*These authors contributed equally to this work,Correspondence to:Jen-Chieh Lee, email:
| | - Shu Liu
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA,*These authors contributed equally to this work,Shu Liu, email:
| | - Yucheng Wang
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA,*These authors contributed equally to this work
| | - You Liang
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - David M. Jablons
- 1Thoracic Oncology, Department of Medicine, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
7
|
Chen Y, Li J, Xu L, Găman MA, Zou Z. The genesis and evolution of acute myeloid leukemia stem cells in the microenvironment: From biology to therapeutic targeting. Cell Death Discov 2022; 8:397. [PMID: 36163119 PMCID: PMC9513079 DOI: 10.1038/s41420-022-01193-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 11/09/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by cytogenetic and genomic alterations. Up to now, combination chemotherapy remains the standard treatment for leukemia. However, many individuals diagnosed with AML develop chemotherapeutic resistance and relapse. Recently, it has been pointed out that leukemic stem cells (LSCs) are the fundamental cause of drug resistance and AML relapse. LSCs only account for a small subpopulation of all leukemic cells, but possess stem cell properties, including a self-renewal capacity and a multi-directional differentiation potential. LSCs reside in a mostly quiescent state and are insensitive to chemotherapeutic agents. When LSCs reside in a bone marrow microenvironment (BMM) favorable to their survival, they engage into a steady, continuous clonal evolution to better adapt to the action of chemotherapy. Most chemotherapeutic drugs can only eliminate LSC-derived clones, reducing the number of leukemic cells in the BM to a normal range in order to achieve complete remission (CR). LSCs hidden in the BM niche can hardly be targeted or eradicated, leading to drug resistance and AML relapse. Understanding the relationship between LSCs, the BMM, and the generation and evolution laws of LSCs can facilitate the development of effective therapeutic targets and increase the efficiency of LSCs elimination in AML.
Collapse
Affiliation(s)
- Yongfeng Chen
- Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, Zhejiang, 318000, China.
| | - Jing Li
- Department of Histology and Embryology, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Linglong Xu
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, China
| | - Mihnea-Alexandru Găman
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 050474, Bucharest, Romania.
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest, Romania.
| | - Zhenyou Zou
- Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou, Guangxi, 545005, China.
| |
Collapse
|
8
|
Zhai Y, Singh P, Dolnik A, Brazda P, Atlasy N, del Gaudio N, Döhner K, Döhner H, Minucci S, Martens J, Altucci L, Megchelenbrink W, Bullinger L, Stunnenberg HG. Longitudinal single-cell transcriptomics reveals distinct patterns of recurrence in acute myeloid leukemia. Mol Cancer 2022; 21:166. [PMID: 35986270 PMCID: PMC9389773 DOI: 10.1186/s12943-022-01635-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/07/2022] [Indexed: 12/02/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a heterogeneous and aggressive blood cancer that results from diverse genetic aberrations in the hematopoietic stem or progenitor cells (HSPCs) leading to the expansion of blasts in the hematopoietic system. The heterogeneity and evolution of cancer blasts can render therapeutic interventions ineffective in a yet poorly understood patient-specific manner. In this study, we investigated the clonal heterogeneity of diagnosis (Dx) and relapse (Re) pairs at genetic and transcriptional levels, and unveiled the underlying pathways and genes contributing to recurrence. Methods Whole-exome sequencing was used to detect somatic mutations and large copy number variations (CNVs). Single cell RNA-seq was performed to investigate the clonal heterogeneity between Dx-Re pairs and amongst patients. Results scRNA-seq analysis revealed extensive expression differences between patients and Dx-Re pairs, even for those with the same -presumed- initiating events. Transcriptional differences between and within patients are associated with clonal composition and evolution, with the most striking differences in patients that gained large-scale copy number variations at relapse. These differences appear to have significant molecular implications, exemplified by a DNMT3A/FLT3-ITD patient where the leukemia switched from an AP-1 regulated clone at Dx to a mTOR signaling driven clone at Re. The two distinct AML1-ETO pairs share genes related to hematopoietic stem cell maintenance and cell migration suggesting that the Re leukemic stem cell-like (LSC-like) cells evolved from the Dx cells. Conclusions In summary, the single cell RNA data underpinned the tumor heterogeneity not only amongst patient blasts with similar initiating mutations but also between each Dx-Re pair. Our results suggest alternatively and currently unappreciated and unexplored mechanisms leading to therapeutic resistance and AML recurrence. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-022-01635-4.
Collapse
|
9
|
Zhai Y, Singh P, Dolnik A, Brazda P, Atlasy N, Del Gaudio N, Döhner K, Döhner H, Minucci S, Martens J, Altucci L, Megchelenbrink W, Bullinger L, Stunnenberg HG. Longitudinal single-cell transcriptomics reveals distinct patterns of recurrence in acute myeloid leukemia. Mol Cancer 2022. [PMID: 35986270 DOI: 10.1186/s12943-022-01635-4.pmid:35986270;pmcid:pmc9389773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous and aggressive blood cancer that results from diverse genetic aberrations in the hematopoietic stem or progenitor cells (HSPCs) leading to the expansion of blasts in the hematopoietic system. The heterogeneity and evolution of cancer blasts can render therapeutic interventions ineffective in a yet poorly understood patient-specific manner. In this study, we investigated the clonal heterogeneity of diagnosis (Dx) and relapse (Re) pairs at genetic and transcriptional levels, and unveiled the underlying pathways and genes contributing to recurrence. METHODS Whole-exome sequencing was used to detect somatic mutations and large copy number variations (CNVs). Single cell RNA-seq was performed to investigate the clonal heterogeneity between Dx-Re pairs and amongst patients. RESULTS scRNA-seq analysis revealed extensive expression differences between patients and Dx-Re pairs, even for those with the same -presumed- initiating events. Transcriptional differences between and within patients are associated with clonal composition and evolution, with the most striking differences in patients that gained large-scale copy number variations at relapse. These differences appear to have significant molecular implications, exemplified by a DNMT3A/FLT3-ITD patient where the leukemia switched from an AP-1 regulated clone at Dx to a mTOR signaling driven clone at Re. The two distinct AML1-ETO pairs share genes related to hematopoietic stem cell maintenance and cell migration suggesting that the Re leukemic stem cell-like (LSC-like) cells evolved from the Dx cells. CONCLUSIONS In summary, the single cell RNA data underpinned the tumor heterogeneity not only amongst patient blasts with similar initiating mutations but also between each Dx-Re pair. Our results suggest alternatively and currently unappreciated and unexplored mechanisms leading to therapeutic resistance and AML recurrence.
Collapse
Affiliation(s)
- Yanan Zhai
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naples, Italy.,Prinses Maxima Centrum, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.,Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands
| | - Prashant Singh
- Prinses Maxima Centrum, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Anna Dolnik
- Medical Department, Division of Hematology, Oncology, and Cancer Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Brazda
- Prinses Maxima Centrum, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.,Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands
| | - Nader Atlasy
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands
| | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naples, Italy
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Saverio Minucci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, EO, Italy
| | - Joost Martens
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naples, Italy.,Institute of Molecular Biology and Genetics, BIOGEM, Ariano Irpino, AV, Italy
| | - Wout Megchelenbrink
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138, Naples, Italy.,Prinses Maxima Centrum, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Lars Bullinger
- Medical Department, Division of Hematology, Oncology, and Cancer Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hendrik G Stunnenberg
- Prinses Maxima Centrum, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands. .,Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
10
|
Barreto IV, Pessoa FMCDP, Machado CB, Pantoja LDC, Ribeiro RM, Lopes GS, Amaral de Moraes ME, de Moraes Filho MO, de Souza LEB, Burbano RMR, Khayat AS, Moreira-Nunes CA. Leukemic Stem Cell: A Mini-Review on Clinical Perspectives. Front Oncol 2022; 12:931050. [PMID: 35814466 PMCID: PMC9270022 DOI: 10.3389/fonc.2022.931050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are known for their ability to proliferate and self-renew, thus being responsible for sustaining the hematopoietic system and residing in the bone marrow (BM). Leukemic stem cells (LSCs) are recognized by their stemness features such as drug resistance, self-renewal, and undifferentiated state. LSCs are also present in BM, being found in only 0.1%, approximately. This makes their identification and even their differentiation difficult since, despite the mutations, they are cells that still have many similarities with HSCs. Although the common characteristics, LSCs are heterogeneous cells and have different phenotypic characteristics, genetic mutations, and metabolic alterations. This whole set of alterations enables the cell to initiate the process of carcinogenesis, in addition to conferring drug resistance and providing relapses. The study of LSCs has been evolving and its application can help patients, where through its count as a biomarker, it can indicate a prognostic factor and reveal treatment results. The selection of a target to LSC therapy is fundamental. Ideally, the target chosen should be highly expressed by LSCs, highly selective, absence of expression on other cells, in particular HSC, and preferentially expressed by high numbers of patients. In view of the large number of similarities between LSCs and HSCs, it is not surprising that current treatment approaches are limited. In this mini review we seek to describe the immunophenotypic characteristics and mechanisms of resistance presented by LSCs, also approaching possible alternatives for the treatment of patients.
Collapse
Affiliation(s)
- Igor Valentim Barreto
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Flávia Melo Cunha de Pinho Pessoa
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Caio Bezerra Machado
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Laudreísa da Costa Pantoja
- Department of Pediatrics, Octávio Lobo Children’s Hospital, Belém, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
| | | | | | - Maria Elisabete Amaral de Moraes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Manoel Odorico de Moraes Filho
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | | | | | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
| | - Caroline Aquino Moreira-Nunes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
- Ceará State University, Northeast Biotechnology Network (RENORBIO), Fortaleza, Brazil
- *Correspondence: Caroline Aquino Moreira-Nunes,
| |
Collapse
|
11
|
Nirachonkul W, Ogonoki S, Thumvijit T, Chiampanichayakul S, Panyajai P, Anuchapreeda S, Tima S, Chiampanichayakul S. CD123-Targeted Nano-Curcumin Molecule Enhances Cytotoxic Efficacy in Leukemic Stem Cells. NANOMATERIALS 2021; 11:nano11112974. [PMID: 34835741 PMCID: PMC8620973 DOI: 10.3390/nano11112974] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/18/2022]
Abstract
Acute myeloblastic leukemia (AML) is a disease with a high rate of relapse and drug resistance due to the remaining leukemic stem cells (LSCs). Therefore, LSCs are specific targets for the treatment of leukemia. CD123 is specifically expressed on LSCs and performs as a specific marker. Curcumin is the main active compound of a natural product with low toxicity for humans. It has been reported to inhibit leukemic cell growth. However, curcumin is practically insoluble in water and has low bioavailability. In this study, we aimed to formulate curcumin nanoparticles and conjugate with the anti-CD123 to overcome the low water solubility and improve the targeting of LSCs. The cytotoxicity of both curcumin-loaded PLGA/poloxamer nanoparticles (Cur-NPs) and anti-CD123-curcumin-loaded PLGA/poloxamer nanoparticles (anti-CD123-Cur-NPs) were examined in KG-1a cells. The results showed that Cur-NPs and Cur-NPs-CD123 exhibited cytotoxic effects on KG-1a cells with the IC50 values of 74.20 ± 6.71 and 41.45 ± 5.49 µM, respectively. Moreover, anti-CD123-Cur-NPs induced higher apoptosis than Cur-NPs. The higher uptake of anti-CD123-Cur-NPs in KG-1a cells was confirmed by using flow cytometry. In conclusion, the anti-CD123-Cur-NPs formulation improved curcumin's bioavailability and specific targeting of LSCs, suggesting that it is a promising drug delivery system for improving the therapeutic efficacy against AML.
Collapse
Affiliation(s)
- Wariya Nirachonkul
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (W.N.); (P.P.); (S.A.); (S.T.)
| | - Siriporn Ogonoki
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
- Research Center of Pharmaceutical Nanotechnology, Faculty Chiang Mai University, Chiang Mai 50200, Thailand
| | - Tarika Thumvijit
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
- Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Pawaret Panyajai
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (W.N.); (P.P.); (S.A.); (S.T.)
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (W.N.); (P.P.); (S.A.); (S.T.)
- Research Center of Pharmaceutical Nanotechnology, Faculty Chiang Mai University, Chiang Mai 50200, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (W.N.); (P.P.); (S.A.); (S.T.)
- Research Center of Pharmaceutical Nanotechnology, Faculty Chiang Mai University, Chiang Mai 50200, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sawitree Chiampanichayakul
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (W.N.); (P.P.); (S.A.); (S.T.)
- Research Center of Pharmaceutical Nanotechnology, Faculty Chiang Mai University, Chiang Mai 50200, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: ; Tel.: +66-5394-9237
| |
Collapse
|
12
|
Thoms JAI, Truong P, Subramanian S, Knezevic K, Harvey G, Huang Y, Seneviratne JA, Carter DR, Joshi S, Skhinas J, Chacon D, Shah A, de Jong I, Beck D, Göttgens B, Larsson J, Wong JWH, Zanini F, Pimanda JE. Disruption of a GATA2-TAL1-ERG regulatory circuit promotes erythroid transition in healthy and leukemic stem cells. Blood 2021; 138:1441-1455. [PMID: 34075404 DOI: 10.1182/blood.2020009707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/03/2021] [Indexed: 10/21/2022] Open
Abstract
Changes in gene regulation and expression govern orderly transitions from hematopoietic stem cells to terminally differentiated blood cell types. These transitions are disrupted during leukemic transformation, but knowledge of the gene regulatory changes underpinning this process is elusive. We hypothesized that identifying core gene regulatory networks in healthy hematopoietic and leukemic cells could provide insights into network alterations that perturb cell state transitions. A heptad of transcription factors (LYL1, TAL1, LMO2, FLI1, ERG, GATA2, and RUNX1) bind key hematopoietic genes in human CD34+ hematopoietic stem and progenitor cells (HSPCs) and have prognostic significance in acute myeloid leukemia (AML). These factors also form a densely interconnected circuit by binding combinatorially at their own, and each other's, regulatory elements. However, their mutual regulation during normal hematopoiesis and in AML cells, and how perturbation of their expression levels influences cell fate decisions remains unclear. In this study, we integrated bulk and single-cell data and found that the fully connected heptad circuit identified in healthy HSPCs persists, with only minor alterations in AML, and that chromatin accessibility at key heptad regulatory elements was predictive of cell identity in both healthy progenitors and leukemic cells. The heptad factors GATA2, TAL1, and ERG formed an integrated subcircuit that regulates stem cell-to-erythroid transition in both healthy and leukemic cells. Components of this triad could be manipulated to facilitate erythroid transition providing a proof of concept that such regulatory circuits can be harnessed to promote specific cell-type transitions and overcome dysregulated hematopoiesis.
Collapse
Affiliation(s)
| | - Peter Truong
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Shruthi Subramanian
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Kathy Knezevic
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Gregory Harvey
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Yizhou Huang
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Janith A Seneviratne
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Daniel R Carter
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Swapna Joshi
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Joanna Skhinas
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Diego Chacon
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Anushi Shah
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Ineke de Jong
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Dominik Beck
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Berthold Göttgens
- Wellcome and Medical Research Council (MRC) Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Jonas Larsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jason W H Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Fabio Zanini
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia; and
| | - John E Pimanda
- School of Medical Sciences
- Adult Cancer Program, and
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
- Department of Haematology, Prince of Wales Hospital, Randwick, NSW, Australia
| |
Collapse
|
13
|
Song J, Shang B, Pei Y, Shi M, Niu X, Dou L, Drokow EK, Xu F, Bai Y, Sun K. A higher percentage of leukemic blasts with vacuoles predicts unfavorable outcomes in patients with acute myeloid leukemia. Leuk Res 2021; 109:106638. [PMID: 34116372 DOI: 10.1016/j.leukres.2021.106638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/14/2021] [Accepted: 06/01/2021] [Indexed: 10/21/2022]
Abstract
Cytoplasmic vacuoles, which are a morphological feature of dysplasia, can be observed under a microscope at initial diagnosis. Recently, this typical morphological feature has been found to be associated with impaired survival. To investigate the clinical significance of the grading of blasts with vacuoles in acute myeloid leukemia (AML), we retrospectively studied 152 patients newly diagnosed with non-M3 AML. The patients were categorized into three groups according to the percentage of blasts with vacuoles (>20 %, 11-20 %, 0-10 %). A high percentage of blasts with vacuoles (>20 %) was positively associated with the European Leukemia Net (2017-ELN) high-risk AML, a complex karyotype, TP53 and IDH1/2 mutations, and CD71 expression and negatively associated with the ELN low-risk category. Importantly, patients who had a higher percentage of blasts with vacuoles had a lower complete remission rate in response to first-cycle induction chemotherapy. The overall survival and event-free survival of patients who had a higher percentage of blasts with vacuoles were significantly shorter. Moreover, multivariate analysis showed that blast vacuolization was an independent high prognostic factor for AML. In conclusion, a higher percentage of leukemic blasts with vacuoles predicts worse outcomes in AML and may have potential as a prognostic marker.
Collapse
Affiliation(s)
- Juanjuan Song
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Baojun Shang
- Institute of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Yanru Pei
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Mingyue Shi
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Xiaona Niu
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Liurui Dou
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Emmanuel Kwateng Drokow
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Fangfang Xu
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Yanliang Bai
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China
| | - Kai Sun
- Department of Hematology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Henan, People's Republic of China.
| |
Collapse
|
14
|
Dembitz V, Gallipoli P. The Role of Metabolism in the Development of Personalized Therapies in Acute Myeloid Leukemia. Front Oncol 2021; 11:665291. [PMID: 34094959 PMCID: PMC8170311 DOI: 10.3389/fonc.2021.665291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/26/2021] [Indexed: 01/17/2023] Open
Abstract
Despite significant recent advances in our understanding of the biology and genetics of acute myeloid leukemia (AML), current AML therapies are mostly based on a backbone of standard chemotherapy which has remained mostly unchanged for over 20 years. Several novel therapies, mostly targeting neomorphic/activating recurrent mutations found in AML patients, have only recently been approved following encouraging results, thus providing the first evidence of a more precise and personalized approach to AML therapy. Rewired metabolism has been described as a hallmark of cancer and substantial evidence of its role in AML establishment and maintenance has been recently accrued in preclinical models. Interestingly, unique metabolic changes are generated by specific AML recurrent mutations or in response to diverse AML therapies, thus creating actionable metabolic vulnerabilities in specific patient groups. In this review we will discuss the current evidence supporting a role for rewired metabolism in AML pathogenesis and how these metabolic changes can be leveraged to develop novel personalized therapies.
Collapse
Affiliation(s)
| | - Paolo Gallipoli
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
15
|
Takahashi M, Barile M, Chapple RH, Tseng YJ, Nakada D, Busch K, Fanti AK, Säwén P, Bryder D, Höfer T, Göttgens B. Reconciling Flux Experiments for Quantitative Modeling of Normal and Malignant Hematopoietic Stem/Progenitor Dynamics. Stem Cell Reports 2021; 16:741-753. [PMID: 33770496 PMCID: PMC8072066 DOI: 10.1016/j.stemcr.2021.02.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Hematopoiesis serves as a paradigm for how homeostasis is maintained within hierarchically organized cell populations. However, important questions remain as to the contribution of hematopoietic stem cells (HSCs) toward maintaining steady state hematopoiesis. A number of in vivo lineage labeling and propagation studies have given rise to contradictory interpretations, leaving key properties of stem cell function unresolved. Using processed flow cytometry data coupled with a biology-driven modeling approach, we show that in vivo flux experiments that come from different laboratories can all be reconciled into a single unifying model, even though they had previously been interpreted as being contradictory. We infer from comparative analysis that different transgenic models display distinct labeling efficiencies across a heterogeneous HSC pool, which we validate by marker gene expression associated with HSC function. Finally, we show how the unified model of HSC differentiation can be used to simulate clonal expansion in the early stages of leukemogenesis.
Collapse
Affiliation(s)
- Munetomo Takahashi
- Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge CB2 0AW, UK; Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Melania Barile
- Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge CB2 0AW, UK.
| | - Richard H Chapple
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yu-Jung Tseng
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Daisuke Nakada
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Katrin Busch
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ann-Kathrin Fanti
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petter Säwén
- Division for Molecular Hematology at Institute for Experimental Medical Sciences, Lund University, Lund, Sweden
| | - David Bryder
- Department of Microbiology and Immunology at the Institute of Biomedicine, Göteborg, Sweden
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Berthold Göttgens
- Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge CB2 0AW, UK.
| |
Collapse
|
16
|
Rodrigues ACBDC, Costa RGA, Silva SLR, Dias IRSB, Dias RB, Bezerra DP. Cell signaling pathways as molecular targets to eliminate AML stem cells. Crit Rev Oncol Hematol 2021; 160:103277. [PMID: 33716201 DOI: 10.1016/j.critrevonc.2021.103277] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/25/2021] [Accepted: 02/27/2021] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) remains the most lethal of leukemias and a small population of cells called leukemic stem cells (LSCs) has been associated with disease relapses. Some cell signaling pathways play an important role in AML survival, proliferation and self-renewal properties and are abnormally activated or suppressed in LSCs. This includes the NF-κB, Wnt/β-catenin, Hedgehog, Notch, EGFR, JAK/STAT, PI3K/AKT/mTOR, TGF/SMAD and PPAR pathways. This review aimed to discuss these pathways as molecular targets for eliminating AML LSCs. Herein, inhibitors/activators of these pathways were summarized as a potential new anti-AML therapy capable of eliminating LSCs to guide future researches. The clinical use of cell signaling pathways data can be useful to enhance the anti-AML therapy.
Collapse
Affiliation(s)
| | - Rafaela G A Costa
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Suellen L R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Ingrid R S B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
17
|
Swatler J, Turos-Korgul L, Kozlowska E, Piwocka K. Immunosuppressive Cell Subsets and Factors in Myeloid Leukemias. Cancers (Basel) 2021; 13:cancers13061203. [PMID: 33801964 PMCID: PMC7998753 DOI: 10.3390/cancers13061203] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Effector immune system cells have the ability to kill tumor cells. However, as a cancer (such as leukemia) develops, it inhibits and evades the effector immune response. Such a state of immunosuppression can be driven by several factors – receptors, soluble cytokines, as well as by suppressive immune cells. In this review, we describe factors and cells that constitute immunosuppressive microenvironment of myeloid leukemias. We characterize factors of direct leukemic origin, such as inhibitory receptors, enzymes and extracellular vesicles. Furthermore, we describe suppressive immune cells, such as myeloid derived suppressor cells and regulatory T cells. Finally, we sum up changes in these drivers of immune evasion in myeloid leukemias during therapy. Abstract Both chronic myeloid leukemia and acute myeloid leukemia evade the immune response during their development and disease progression. As myeloid leukemia cells modify their bone marrow microenvironment, they lead to dysfunction of cytotoxic cells, such as CD8+ T cells or NK cells, simultaneously promoting development of immunosuppressive regulatory T cells and suppressive myeloid cells. This facilitates disease progression, spreading of leukemic blasts outside the bone marrow niche and therapy resistance. The following review focuses on main immunosuppressive features of myeloid leukemias. Firstly, factors derived directly from leukemic cells – inhibitory receptors, soluble factors and extracellular vesicles, are described. Further, we outline function, properties and origin of main immunosuppressive cells - regulatory T cells, myeloid derived suppressor cells and macrophages. Finally, we analyze interplay between recovery of effector immunity and therapeutic modalities, such as tyrosine kinase inhibitors and chemotherapy.
Collapse
Affiliation(s)
- Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
| | - Laura Turos-Korgul
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
| | - Ewa Kozlowska
- Department of Immunology, Institute of Functional Biology and Ecology, University of Warsaw, 02-096 Warsaw, Poland;
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
- Correspondence:
| |
Collapse
|
18
|
Stahlhut M, Ha TC, Takmakova E, Morgan MA, Schwarzer A, Schaudien D, Eder M, Schambach A, Kustikova OS. Conditionally immortalised leukaemia initiating cells co-expressing Hoxa9/Meis1 demonstrate microenvironmental adaptation properties ex vivo while maintaining myelomonocytic memory. Sci Rep 2021; 11:5294. [PMID: 33674652 PMCID: PMC7935976 DOI: 10.1038/s41598-021-84468-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 02/12/2021] [Indexed: 01/31/2023] Open
Abstract
Regulation of haematopoietic stem cell fate through conditional gene expression could improve understanding of healthy haematopoietic and leukaemia initiating cell (LIC) biology. We established conditionally immortalised myeloid progenitor cell lines co-expressing constitutive Hoxa9.EGFP and inducible Meis1.dTomato (H9M-ciMP) to study growth behaviour, immunophenotype and morphology under different cytokine/microenvironmental conditions ex vivo upon doxycycline (DOX) induction or removal. The vector design and drug-dependent selection approach identified new retroviral insertion (RVI) sites that potentially collaborate with Meis1/Hoxa9 and define H9M-ciMP fate. For most cell lines, myelomonocytic conditions supported reversible H9M-ciMP differentiation into neutrophils and macrophages with DOX-dependent modulation of Hoxa9/Meis1 and CD11b/Gr-1 expression. Here, up-regulation of Meis1/Hoxa9 promoted reconstitution of exponential expansion of immature H9M-ciMPs after DOX reapplication. Stem cell maintaining conditions supported selective H9M-ciMP exponential growth. H9M-ciMPs that had Ninj2 RVI and were cultured under myelomonocytic or stem cell maintaining conditions revealed the development of DOX-dependent acute myeloid leukaemia in a murine transplantation model. Transcriptional dysregulation of Ninj2 and distal genes surrounding RVI (Rad52, Kdm5a) was detected. All studied H9M-ciMPs demonstrated adaptation to T-lymphoid microenvironmental conditions while maintaining immature myelomonocytic features. Thus, the established system is relevant to leukaemia and stem cell biology.
Collapse
Affiliation(s)
- Maike Stahlhut
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Teng Cheong Ha
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Ekaterina Takmakova
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Michael A Morgan
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Adrian Schwarzer
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Matthias Eder
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany.
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Olga S Kustikova
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany.
- REBIRTH - Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
19
|
Lopez-Reyes RG, Quinet G, Gonzalez-Santamarta M, Larrue C, Sarry JE, Rodriguez MS. Inhibition of the proteasome and proteaphagy enhances apoptosis in FLT3-ITD-driven acute myeloid leukemia. FEBS Open Bio 2020; 11:48-60. [PMID: 33410599 PMCID: PMC7780102 DOI: 10.1002/2211-5463.12950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/15/2020] [Accepted: 08/12/2020] [Indexed: 12/05/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a clonal disorder that affects hematopoietic stem cells or myeloid progenitors. One of the most common mutations that results in AML occurs in the gene encoding fms‐like tyrosine kinase 3 (FLT3). Previous studies have demonstrated that AML cells expressing FLT3‐internal tandem duplication (ITD) are more sensitive to the proteasome inhibitor bortezomib (Bz) than FLT3 wild‐type cells, with this cytotoxicity being mediated by autophagy (Atg). Here, we show that proteasome inhibition with Bz results in modest but consistent proteaphagy in MOLM‐14 leukemic cells expressing the FLT3‐ITD mutation, but not in OCI‐AML3 leukemic cells with wild‐type FLT3. Chemical inhibition of Atg with bafilomycin A simultaneously blocked proteaphagy and resulted in the accumulation of the p62 Atg receptor in Bz‐treated MOLM‐14 cells. The use of ubiquitin traps revealed that ubiquitin plays an important role in proteasome‐Atg cross‐talk. The p62 inhibitor verteporfin blocked proteaphagy and, importantly, resulted in accumulation of high molecular weight forms of p62 and FLT3‐ITD in Bz‐treated MOLM‐14 cells. Both Atg inhibitors enhanced Bz‐induced apoptosis in FLT3‐ITD‐driven leukemic cells, highlighting the therapeutic potential of these treatments.
Collapse
Affiliation(s)
- Rosa G Lopez-Reyes
- Institute of Advanced Technology and Life Sciences (ITAV), IPBS-Centre de la Recherche Scientifique (CNRS), Université Toulouse III Paul Sabatier, Toulouse, France.,Cancer Research Center of Toulouse Unité Mixte de Recherche (UMR) 1037 INSERM, ERL 5294 Centre de la Recherche Scientifique (CNRS), Toulouse, France
| | - Grégoire Quinet
- Institute of Advanced Technology and Life Sciences (ITAV), IPBS-Centre de la Recherche Scientifique (CNRS), Université Toulouse III Paul Sabatier, Toulouse, France
| | - Maria Gonzalez-Santamarta
- Institute of Advanced Technology and Life Sciences (ITAV), IPBS-Centre de la Recherche Scientifique (CNRS), Université Toulouse III Paul Sabatier, Toulouse, France
| | - Clément Larrue
- Cancer Research Center of Toulouse Unité Mixte de Recherche (UMR) 1037 INSERM, ERL 5294 Centre de la Recherche Scientifique (CNRS), Toulouse, France
| | - Jean-Emmanuel Sarry
- Cancer Research Center of Toulouse Unité Mixte de Recherche (UMR) 1037 INSERM, ERL 5294 Centre de la Recherche Scientifique (CNRS), Toulouse, France
| | - Manuel S Rodriguez
- Institute of Advanced Technology and Life Sciences (ITAV), IPBS-Centre de la Recherche Scientifique (CNRS), Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
20
|
Oswald JT, Patel H, Khan D, Jeorje NN, Golzar H, Oswald EL, Tang S. Drug Delivery Systems Using Surface Markers for Targeting Cancer Stem Cells. Curr Pharm Des 2020; 26:2057-2071. [PMID: 32250211 DOI: 10.2174/1381612826666200406084900] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022]
Abstract
The innate abilities of cancer stem cells (CSCs), such as multi-drug resistance, drug efflux, quiescence and ionizing radiation tolerance, protect them from most traditional chemotherapeutics. As a result, this small subpopulation of persistent cells leads to more aggressive and chemoresistant cancers, causing tumour relapse and metastasis. This subpopulation is differentiated from the bulk tumour population through a wide variety of surface markers expressed on the cell surface. Recent developments in nanomedicine and targeting delivery methods have given rise to new possibilities for specifically targeting these markers and preferentially eliminating CSCs. Herein, we first summarize the range of surface markers identifying CSC populations in a variety of cancers; then, we discuss recent attempts to actively target CSCs and their niches using liposomal, nanoparticle, carbon nanotube and viral formulations.
Collapse
Affiliation(s)
- James T Oswald
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Haritosh Patel
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Daid Khan
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Ninweh N Jeorje
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Hossein Golzar
- Department of Chemistry & Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Erin L Oswald
- School Of Nanotechnology Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Shirley Tang
- Department of Chemistry & Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
21
|
Huang Y, Xue X, Li X, Jia B, Pan CX, Li Y, Lin TY. Novel nanococktail of a dual PI3K/mTOR inhibitor and cabazitaxel for castration-resistant prostate cancer. ADVANCED THERAPEUTICS 2020; 3:2000075. [PMID: 33072858 PMCID: PMC7567330 DOI: 10.1002/adtp.202000075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Indexed: 01/09/2023]
Abstract
Prognosis of castration-resistant prostate cancer (CRPC) carries is poor, and no effective therapeutic regimen is yet known. The phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway played a predominant role and may be a promising molecular target for CRPC. However, the toxicity of the dual PI3K inhibitors in clinical trials limits their clinical efficacy for CRPC. To solve this problem, we employed a highly integrated precision nanomedicine strategy to molecularly and physically target CRPC through synergistic effects, enhanced targeted drug delivery efficiency, and reduced unwanted side-effects. Gedatolisib (Ge), a potent inhibitor of PI3K/mTOR, was formulated into our disulfied-crosslinked micelle plateform (NanoGe), which exhibits excellent water solubility, small size (23.25±2 nm), excellent stability with redox stimulus-responsive disintegration, and preferential uptake at tumor sites. NanoGe improved the anti-neoplastic effect of free Ge by 53 times in PC-3M cells and 13 times in C4-2B cells though its enhanced uptake via caveolae- and clathrin-mediated endocytic pathways and the subsequent inhibition of the PI3K/mTOR pathway, resulting in Bax/Bcl-2 dependent apoptosis. In an animal xenograft model, NanoGe showed superior efficacy than free Ge, and synergized with nanoformulated cabazitaxel (NanoCa) as a nanococktail format to achieve a cure rate of 83%. Taken together, our results demonstrate the potency of NanoGe in combination with NanoCa is potent against prostate cancer.
Collapse
Affiliation(s)
- Yee Huang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, P.R. China
| | - Xiangdong Xue
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento CA 95817
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento CA 95817
| | - Bei Jia
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento CA 95817
| | - Chong-xian Pan
- Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento CA 95817
- VA Northern California Health Care System, Mather, CA 95655
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento CA 95817
| | - Tzu-yin Lin
- Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento CA 95817
| |
Collapse
|
22
|
Carrelha J, Lin DS, Rodriguez-Fraticelli AE, Luis TC, Wilkinson AC, Cabezas-Wallscheid N, Tremblay CS, Haas S. Single-cell lineage tracing approaches in hematology research: technical considerations. Exp Hematol 2020; 89:26-36. [PMID: 32735908 PMCID: PMC7894992 DOI: 10.1016/j.exphem.2020.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 01/27/2023]
Abstract
The coordinated differentiation of hematopoietic stem and progenitor cells (HSPCs) into the various mature blood cell types is responsible for sustaining blood and immune system homeostasis. The cell fate decisions underlying this important biological process are made at the level of single cells. Methods to trace the fate of single cells are therefore essential for understanding hematopoietic system activity in health and disease and have had a major impact on how we understand and represent hematopoiesis. Here, we discuss the basic methodologies and technical considerations for three important clonal assays: single-cell transplantation, lentiviral barcoding, and Sleeping Beauty barcoding. This perspective is a synthesis of presentations and discussions from the 2019 International Society for Experimental Hematology (ISEH) Annual Meeting New Investigator Technology Session and the 2019 ISEH Winter Webinar.
Collapse
Affiliation(s)
- Joana Carrelha
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK; MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Dawn S Lin
- Immunology, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Alejo E Rodriguez-Fraticelli
- Stem Cell Program, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Tiago C Luis
- Department of Life Sciences, Imperial College London, London, UK
| | - Adam C Wilkinson
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Cedric S Tremblay
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
| |
Collapse
|
23
|
Shahid S, Shakeel M, Siddiqui S, Ahmed S, Sohail M, Khan IA, Abid A, Shamsi T. Novel Genetic Variations in Acute Myeloid Leukemia in Pakistani Population. Front Genet 2020; 11:560. [PMID: 32655615 PMCID: PMC7324646 DOI: 10.3389/fgene.2020.00560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 05/07/2020] [Indexed: 12/22/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by clonal expansion of blast cells that exhibit great genetic heterogeneity. In this study, we describe the mutational landscape and its clinico-pathological significance in 26 myeloid neoplasm patients from a South Asian population (Pakistan) by using ultra-deep targeted next-generation DNA sequencing of 54 genes (∼5000×) and its subsequent bioinformatics analysis. The data analysis indicated novel non-silent somatic mutational events previously not reported in AML, including nine non-synonymous and one stop-gain mutations. Notably, two recurrent somatic non-synonymous mutations, i.e., STAG2 (causing p.L526F) and BCORL1 (p.A400V), were observed in three unrelated cases each. The BCOR was found to have three independent non-synonymous somatic mutations in three cases. Further, the SRSF2 with a protein truncating somatic mutation (p.Q88X) was observed for the first time in AML in this study. The prioritization of germline mutations with ClinVar, SIFT, Polyphen2, and Combined Annotation Dependent Depletion (CADD) highlighted 18 predicted deleterious/pathogenic mutations, including two recurrent deleterious mutations, i.e., a novel heterozygous non-synonymous SNV in GATA2 (p.T358P) and a frameshift insertion in NPM1 (p.L258fs), found in two unrelated cases each. The WT1 was observed with three independent potential detrimental germline mutations in three different cases. Collectively, non-silent somatic and/or germline mutations were observed in 23 (88.46%) of the cases (0.92 mutation per case). Furthermore, the pharmGKB database exploration showed a missense SNV rs1042522 in TP53, exhibiting decreased response to anti-cancer drugs, in 19 (73%) of the cases. This genomic profiling of AML provides deep insight into the disease pathophysiology. Identification of pharmacogenomics markers will help to adopt personalized approach for the management of AML patients in Pakistan.
Collapse
Affiliation(s)
- Saba Shahid
- Department of Genomics, National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Muhammad Shakeel
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Saima Siddiqui
- Department of Hematology, National Institute of Blood Diseases and Bone Marrow Transplantation Karachi, Karachi, Pakistan
| | - Shariq Ahmed
- Department of Genomics, National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Misha Sohail
- Department of Genomics, National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Ishtiaq Ahmad Khan
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Aiysha Abid
- Centre for Human Genetics and Molecular Medicine, Sindh Institute of Urology and Transplantation (SIUT), Karachi, Pakistan
| | - Tahir Shamsi
- Department of Hematology, National Institute of Blood Diseases and Bone Marrow Transplantation Karachi, Karachi, Pakistan
| |
Collapse
|
24
|
Yang W, Xie J, Hou R, Chen X, Xu Z, Tan Y, Ren F, Zhang Y, Xu J, Chang J, Wang H. Disulfiram/cytarabine eradicates a subset of acute myeloid leukemia stem cells with high aldehyde dehydrogenase expression. Leuk Res 2020; 92:106351. [PMID: 32224355 DOI: 10.1016/j.leukres.2020.106351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 01/07/2023]
Abstract
Most patients with acute myeloid leukemia (AML) achieve complete remission (CR) after induction chemotherapy, however, in some patients, the disease subsequently relapses and may lead to death. Leukemia stem cells (LSC) have been identified as the main cause for recurrence. Increased aldehyde dehydrogenase (ALDHhigh) activity in a variety of cancer stem cells prevents effective action of chemotherapeutic drugs. In this study, we found that approximately 50.7% of AML patients had ALDHhigh, and the presence of ALDHhigh stem cells was associated with poor cytogenetic prognosis. Lentiviral vector transduced ALDHhigh leukemia cell lines are insensitive to the conventional chemotherapy drug cytarabine, and inhibition of ALDH activity by disulfiram (DSF) can increase the sensitivity of ALDHhigh leukemia cells to cytarabine. Unlike traditional chemotherapy drugs, DSF is not toxic to healthy umbilical cord blood stem cells. An ALDHhigh leukemia cell xenograft model was established using immunodeficient mice to mimic the disease environment, and DSF and cytarabine were found to eliminate the ALDHhigh leukemia cells in transplanted mice while not affecting the healthy blood cells of mice. These findings suggest that DSF may have therapeutic potential by inhibiting ALDH activity and thereby increasing chemosensitivity.
Collapse
Affiliation(s)
- Wanfang Yang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China; Shanxi University of Chinese Medicine, Jinzhong, China
| | - Juan Xie
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruixia Hou
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiuhua Chen
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhifang Xu
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanhong Tan
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Fanggang Ren
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yaofang Zhang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing Xu
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jianmei Chang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongwei Wang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
25
|
Ladikou EE, Sivaloganathan H, Pepper A, Chevassut T. Acute Myeloid Leukaemia in Its Niche: the Bone Marrow Microenvironment in Acute Myeloid Leukaemia. Curr Oncol Rep 2020; 22:27. [PMID: 32048054 PMCID: PMC7012995 DOI: 10.1007/s11912-020-0885-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose of Review Acute myeloid leukaemia (AML) is a heterogeneous malignancy for which treatment options remain suboptimal. It is clear that a greater understanding of the biology of the AML niche will enable new therapeutic strategies to be developed in order to improve treatment outcomes for patients. Recent Findings Recent evidence has highlighted the importance of the bone marrow microenvironment in protecting leukaemia cells, and in particular leukaemic stem cells from chemotherapy-induced cell death. This includes mesenchymal stem cells supporting growth and preventing apoptosis, and altered action and secretion profiles of other niche components including adipocytes, endothelial cells and T cells. Summary Here, we provide a detailed overview of the current understanding of the AML bone marrow microenvironment. Clinical trials of agents that mobilise leukaemic stem cells from the bone marrow are currently ongoing and show early promise. Future challenges will involve combining these novel therapies targeted at the AML niche with conventional chemotherapy treatment.
Collapse
Affiliation(s)
- E E Ladikou
- Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PS, UK.,Royal Sussex County Hospital, Brighton, BN2 5BE, UK
| | - H Sivaloganathan
- Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PS, UK
| | - A Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PS, UK
| | - T Chevassut
- Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PS, UK. .,Royal Sussex County Hospital, Brighton, BN2 5BE, UK.
| |
Collapse
|
26
|
Dao FT, Yang L, Wang YZ, Chang Y, Jiang Q, Jiang H, Liu YR, Huang XJ, Qin YZ. [Characteristic and prognostic significance of leukemia stem cells associated antigens expressions in t (8;21) acute myeloid leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:831-836. [PMID: 31775482 PMCID: PMC7364990 DOI: 10.3760/cma.j.issn.0253-2727.2019.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Indexed: 11/05/2022]
Abstract
Objective: To investigate the characteristic and prognostic significance of leukemia stem cells associated antigens expressions including CD34, CD38, CD123, CD96 and TIM-3 in t (8;21) AML. Methods: Bone marrow samples of 47 t (8;21) AML patients were collected at diagnosis from October 2015 to April 2018 in Peking University Peoples' Hospital, then flow cytometry method was performed to detect the expression frequencies of CD34, CD38, CD123, CD96 and TIM-3 to analyze the relationship between leukemia stem cells associated antigens expressions and relapse. Results: Of 47 t (8;21) AML patients tested, the median percentages of CD34(+)CD38(-), CD34(+) CD38(-)CD123(+), CD34(+)CD38(-) CD96(+) and CD34(+) CD38(-) TIM-3(+) cells among nucleated cells were 2.37%, 0.24%, 0.27% and 0.06%, respectively. All the frequencies of CD34(+)CD38(-), CD34(+)CD38(-)CD123(+), CD34(+)CD38(-)CD96(+) and CD34(+) CD38(-)TIM-3(+) cells had no impact on the achievement of CR after the first course of induction. All higher frequencies of CD34(+)CD38(-), CD34(+)CD38(-)CD123(+), CD34(+)CD38(-)CD96(+) cells were related to higher 2-year CIR rate. Whereas, the frequency of CD34(+) CD38(-) TIM-3(+) cells had no impact on CIR rate. Both high frequency of CD34(+) CD38(-) cells and the high level of minimal residual diseases (patients with <3-log reduction in the RUNX1-RUNX1T1 transcript level after the second consolidation therapy) were independent poor prognostic factors of CIR[P=0.025, HR=6.9 (95%CI 1.3-37.4) ; P=0.031, HR=11.1 (95%CI 1.2-99.2) ]. Conclusion: Different leukemia stem cells associated antigens had distinct prognostic significance in t (8;21) AML. High frequencies of CD34(+) CD38(-), CD34(+) CD38(-) CD123(+) and CD34(+)CD38(-)CD96(+) cells at diagnosis predicted relapse in patients with t (8;21) AML.
Collapse
Affiliation(s)
- F T Dao
- Peking University Peoples'Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chopra M, Bohlander SK. The cell of origin and the leukemia stem cell in acute myeloid leukemia. Genes Chromosomes Cancer 2019; 58:850-858. [PMID: 31471945 DOI: 10.1002/gcc.22805] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 12/19/2022] Open
Abstract
There is experimental and observational evidence that the cells of the leukemic clone in acute myeloid leukemia (AML) have different phenotypes even though they share the same somatic mutations. The organization of the malignant clone in AML has many similarities to normal hematopoiesis, with leukemia stem cells (LSCs) that sustain leukemia and give rise to more differentiated cells. LSCs, similar to normal hematopoietic stem cells (HSCs), are those cells that are able to give rise to a new leukemic clone when transplanted into a recipient. The cell of origin of leukemia (COL) is defined as the normal cell that is able to transform into a leukemia cell. Current evidence suggests that the COL is distinct from the LSC. Here, we will review the current knowledge about LSCs and the COL in AML.
Collapse
Affiliation(s)
- Martin Chopra
- Leukaemia & Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Stefan K Bohlander
- Leukaemia & Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
28
|
Thoms JAI, Beck D, Pimanda JE. Transcriptional networks in acute myeloid leukemia. Genes Chromosomes Cancer 2019; 58:859-874. [PMID: 31369171 DOI: 10.1002/gcc.22794] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
Acute myeloid leukemia (AML) is a complex disease characterized by a diverse range of recurrent molecular aberrations that occur in many different combinations. Components of transcriptional networks are a common target of these aberrations, leading to network-wide changes and deployment of novel or developmentally inappropriate transcriptional programs. Genome-wide techniques are beginning to reveal the full complexity of normal hematopoietic stem cell transcriptional networks and the extent to which they are deregulated in AML, and new understandings of the mechanisms by which AML cells maintain self-renewal and block differentiation are starting to emerge. The hope is that increased understanding of the network architecture in AML will lead to identification of key oncogenic dependencies that are downstream of multiple network aberrations, and that this knowledge will be translated into new therapies that target these dependencies. Here, we review the current state of knowledge of network perturbation in AML with a focus on major mechanisms of transcription factor dysregulation, including mutation, translocation, and transcriptional dysregulation, and discuss how these perturbations propagate across transcriptional networks. We will also review emerging mechanisms of network disruption, and briefly discuss how increased knowledge of network disruption is already being used to develop new therapies.
Collapse
Affiliation(s)
- Julie A I Thoms
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Dominik Beck
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia.,Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - John E Pimanda
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.,Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.,Department of Haematology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
29
|
Kaur M, Drake AC, Hu G, Rudnick S, Chen Q, Phennicie R, Attar R, Nemeth J, Gaudet F, Chen J. Induction and Therapeutic Targeting of Human NPM1c + Myeloid Leukemia in the Presence of Autologous Immune System in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 202:1885-1894. [PMID: 30710044 DOI: 10.4049/jimmunol.1800366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022]
Abstract
Development of targeted cancer therapy requires a thorough understanding of mechanisms of tumorigenesis as well as mechanisms of action of therapeutics. This is challenging because by the time patients are diagnosed with cancer, early events of tumorigenesis have already taken place. Similarly, development of cancer immunotherapies is hampered by a lack of appropriate small animal models with autologous human tumor and immune system. In this article, we report the development of a mouse model of human acute myeloid leukemia (AML) with autologous immune system for studying early events of human leukemogenesis and testing the efficacy of immunotherapeutics. To develop such a model, human hematopoietic stem/progenitor cells (HSPC) are transduced with lentiviruses expressing a mutated form of nucleophosmin (NPM1), referred to as NPM1c. Following engraftment into immunodeficient mice, transduced HSPCs give rise to human myeloid leukemia, whereas untransduced HSPCs give rise to human immune cells in the same mice. The de novo AML, with CD123+ leukemic stem or initiating cells (LSC), resembles NPM1c+ AML from patients. Transcriptional analysis of LSC and leukemic cells confirms similarity of the de novo leukemia generated in mice with patient leukemia and suggests Myc as a co-operating factor in NPM1c-driven leukemogenesis. We show that a bispecific conjugate that binds both CD3 and CD123 eliminates CD123+ LSCs in a T cell-dependent manner both in vivo and in vitro. These results demonstrate the utility of the NPM1c+ AML model with an autologous immune system for studying early events of human leukemogenesis and for evaluating efficacy and mechanism of immunotherapeutics.
Collapse
Affiliation(s)
- Mandeep Kaur
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Adam C Drake
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Guangan Hu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - Qingfeng Chen
- Institute for Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673
| | - Ryan Phennicie
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ricardo Attar
- Janssen Pharmaceuticals, Inc., Springhouse, PA 19477; and
| | - Jeffrey Nemeth
- Janssen Pharmaceuticals, Inc., Springhouse, PA 19477; and
| | | | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139; .,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
30
|
Kogan AA, Lapidus RG, Baer MR, Rassool FV. Exploiting epigenetically mediated changes: Acute myeloid leukemia, leukemia stem cells and the bone marrow microenvironment. Adv Cancer Res 2019; 141:213-253. [PMID: 30691684 DOI: 10.1016/bs.acr.2018.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) derives from the clonal expansion of immature myeloid cells in the bone marrow, and results in the disruption of normal hematopoiesis and subsequent bone marrow failure. The bone marrow microenvironment (BME) and its immune and other supporting cells are regarded to facilitate the survival, differentiation and proliferation of leukemia stem cells (LSCs), which enables AML cells to persist and expand despite treatment. Recent studies have identified epigenetic modifications among AML cells and BME constituents in AML, and have shown that epigenetic therapy can potentially reprogram these alterations. In this review, we summarize the interactions between the BME and LSCs, and discuss changes in how the BME and immune cells interact with AML cells. After describing the epigenetic modifications seen across chromatin, DNA, the BME, and the immune microenvironment, we explore how demethylating agents may reprogram these pathological interactions, and potentially re-sensitize AML cells to treatment.
Collapse
Affiliation(s)
- Aksinija A Kogan
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States; University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Rena G Lapidus
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Maria R Baer
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Feyruz V Rassool
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States; University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States.
| |
Collapse
|
31
|
Bandyopadhyay S, Fowles JS, Yu L, Fisher DAC, Oh ST. Identification of functionally primitive and immunophenotypically distinct subpopulations in secondary acute myeloid leukemia by mass cytometry. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2019; 96:46-56. [PMID: 30426661 PMCID: PMC6343486 DOI: 10.1002/cyto.b.21743] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 09/09/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Background: Mass cytometry (CyTOF) is a powerful tool for analyzing cellular networks at the single cell level. Due to the high-dimensional nature of this approach, analysis algorithms have been developed to visualize and interpret mass cytometry data. In this study, we applied these approaches to a cohort of patients with secondary acute myeloid leukemia (sAML). METHODS We utilized mass cytometry to interrogate localization and intensity of thrombopoietin-mediated intracellular signaling in sAML. Extracellular and intracellular phenotypes were dissected using SPADE, viSNE, and PhenoGraph. RESULTS Healthy controls exhibited highly localized signaling responses largely restricted to the hematopoietic stem/progenitor cell (HSPC) compartment. In contrast, sAML samples contained subpopulations outside the HSPC compartment exhibiting thrombopoietin (TPO) sensitivity comparable to or greater than immunophenotypically defined HSPCs. We employed unsupervised clustering by PhenoGraph to elucidate distinct subpopulations within these heterogeneous samples. One metacluster composed almost exclusively of Lin- CD61+ CD34- CD38- CD45low cells was identified. This subpopulation was not readily identified by established manual gating approaches, and generally exhibited greater STAT phosphorylation in response to TPO stimulation than did Lin- CD61- CD34+ CD38- cells. Lin- CD61+ CD34- CD38- CD45low cells were identified in three additional sAML patients analyzed independently using a manual gating approach based upon PhenoGraph results. Each patient exhibited a similar TPO hypersensitivity to the PhenoGraph metacluster. CONCLUSIONS The identification of this cellular subpopulation highlights the limitations of manual gating in sAML. Our study demonstrates the potential for mass cytometry to elucidate rare subpopulations in highly heterogeneous tumors by utilizing unsupervised high dimensional analysis. © 2018 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Shovik Bandyopadhyay
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana
- Division of Hematology, Washington University School of Medicine, St Louis, Missouri
| | - Jared S Fowles
- Division of Hematology, Washington University School of Medicine, St Louis, Missouri
| | - Liyang Yu
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel A C Fisher
- Division of Hematology, Washington University School of Medicine, St Louis, Missouri
| | - Stephen T Oh
- Division of Hematology, Washington University School of Medicine, St Louis, Missouri
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
32
|
Leukemia Stem Cells in the Pathogenesis, Progression, and Treatment of Acute Myeloid Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:95-128. [DOI: 10.1007/978-981-13-7342-8_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
Histone deacetylase inhibitor targets CD123/CD47-positive cells and reverse chemoresistance phenotype in acute myeloid leukemia. Leukemia 2018; 33:931-944. [PMID: 30291336 DOI: 10.1038/s41375-018-0279-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/15/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023]
Abstract
Chemoresistance may be due to the survival of leukemia stem cells (LSCs) that are quiescent and not responsive to chemotherapy or lie on the intrinsic or acquired resistance of the specific pool of AML cells. Here, we found, among well-established LSC markers, only CD123 and CD47 are correlated with AML cell chemosensitivities across cell lines and patient samples. Further study reveals that percentages of CD123+CD47+ cells significantly increased in chemoresistant lines compared to parental cell lines. However, stemness signature genes are not significantly increased in resistant cells. Instead, gene changes are enriched in cell cycle and cell survival pathways. This suggests CD123 may serve as a biomarker for chemoresistance, but not stemness of AML cells. We further investigated the role of epigenetic factors in regulating the survival of chemoresistant leukemia cells. Epigenetic drugs, especially histone deacetylase inhibitors (HDACis), effectively induced apoptosis of chemoresistant cells. Furthermore, HDACi Romidepsin largely reversed gene expression profile of resistant cells and efficiently targeted and removed chemoresistant leukemia blasts in xenograft AML mouse model. More interestingly, Romidepsin preferentially targets CD123+ cells, while chemotherapy drug Ara-C mainly targeted fast-growing, CD123- cells. Therefore, Romidepsin alone or in combination with Ara-C may be a potential treatment strategy for chemoresistant patients.
Collapse
|
34
|
Med23 serves as a gatekeeper of the myeloid potential of hematopoietic stem cells. Nat Commun 2018; 9:3746. [PMID: 30218073 PMCID: PMC6138688 DOI: 10.1038/s41467-018-06282-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/22/2018] [Indexed: 12/19/2022] Open
Abstract
In response to myeloablative stresses, HSCs are rapidly activated to replenish myeloid progenitors, while maintaining full potential of self-renewal to ensure life-long hematopoiesis. However, the key factors that orchestrate HSC activities during physiological stresses remain largely unknown. Here we report that Med23 controls the myeloid potential of activated HSCs. Ablation of Med23 in hematopoietic system leads to lymphocytopenia. Med23-deficient HSCs undergo myeloid-biased differentiation and lose the self-renewal capacity. Interestingly, Med23-deficient HSCs are much easier to be activated in response to physiological stresses. Mechanistically, Med23 plays essential roles in maintaining stemness genes expression and suppressing myeloid lineage genes expression. Med23 is downregulated in HSCs and Med23 deletion results in better survival under myeloablative stress. Altogether, our findings identify Med23 as a gatekeeper of myeloid potential of HSCs, thus providing unique insights into the relationship among Med23-mediated transcriptional regulations, the myeloid potential of HSCs and HSC activation upon stresses. Hematopoietic stem cells (HSCs) in the bone marrow are quiescent, but are activated in response to stress. Here, the authors show that loss of Med23 leads to greater activation and enhanced myeloid potential of HSCs in response to stress, also Med23 maintains stemness gene expression and suppresses myeloid genes.
Collapse
|
35
|
Hussaini MO, Mirza AS, Komrokji R, Lancet J, Padron E, Song J. Genetic Landscape of Acute Myeloid Leukemia Interrogated by Next-generation Sequencing: A Large Cancer Center Experience. Cancer Genomics Proteomics 2018; 15:121-126. [PMID: 29496691 DOI: 10.21873/cgp.20070] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 11/23/2017] [Accepted: 11/27/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND/AIM Acute myeloid leukemia (AML) represents a heterogeneous disease with varying morphologic, immunophenotypic, and genetic features, along with varying patient outcomes. The genomic tractability of AML makes it amenable for targeted next-generation sequencing (NGS) testing clinically. MATERIALS AND METHODS One hundred eights-seven unique patients with a diagnosis of acute myeloid leukemia between May 2011 and Oct 2014 and with mutational analysis by NGS were included in this study. The distribution of gene mutations was investigated in different subcategories of AML. RESULTS Most patients in this study (n=182) received Genoptix testing (either 5-gene panel or 21-gene panel). In 130/187 (70%) cases, there was an average of 2.3 mutations per case (range=0-7 mutations). We specifically mention mutations in 32 genes, their significance and co-occurrence as detected in different types of AML. CONCLUSION The genetic heterogeneity of AML signifies the importance of taking a personalized-medicine approach to the management of patients with AML.
Collapse
Affiliation(s)
| | - Abu-Sayeef Mirza
- Department of Internal Medicine, University of South Florida, Tampa, FL, U.S.A
| | - Rami Komrokji
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, U.S.A
| | - Jeffrey Lancet
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, U.S.A
| | - Eric Padron
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, U.S.A
| | - Jinming Song
- Department of Hematopathology, Moffitt Cancer Center, Tampa, FL, U.S.A
| |
Collapse
|
36
|
Mesuraca M, Amodio N, Chiarella E, Scicchitano S, Aloisio A, Codispoti B, Lucchino V, Montalcini Y, Bond HM, Morrone G. Turning Stem Cells Bad: Generation of Clinically Relevant Models of Human Acute Myeloid Leukemia through Gene Delivery- or Genome Editing-Based Approaches. Molecules 2018; 23:E2060. [PMID: 30126100 PMCID: PMC6222541 DOI: 10.3390/molecules23082060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/09/2018] [Accepted: 08/14/2018] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML), the most common acute leukemia in the adult, is believed to arise as a consequence of multiple molecular events that confer on primitive hematopoietic progenitors unlimited self-renewal potential and cause defective differentiation. A number of genetic aberrations, among which a variety of gene fusions, have been implicated in the development of a transformed phenotype through the generation of dysfunctional molecules that disrupt key regulatory mechanisms controlling survival, proliferation, and differentiation in normal stem and progenitor cells. Such genetic aberrations can be recreated experimentally to a large extent, to render normal hematopoietic stem cells "bad", analogous to the leukemic stem cells. Here, we wish to provide a brief outline of the complementary experimental approaches, largely based on gene delivery and more recently on gene editing, employed over the last two decades to gain insights into the molecular mechanisms underlying AML development and progression and on the prospects that their applications offer for the discovery and validation of innovative therapies.
Collapse
Affiliation(s)
- Maria Mesuraca
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Nicola Amodio
- Laboratory of Medical Oncology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Emanuela Chiarella
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Stefania Scicchitano
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Annamaria Aloisio
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Bruna Codispoti
- Tecnologica Research Institute-Marrelli Hospital, 88900 Crotone, Italy.
| | - Valeria Lucchino
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| | - Ylenia Montalcini
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Heather M Bond
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| | - Giovanni Morrone
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, University Magna Græcia, 88100 Catanzaro, Italy.
| |
Collapse
|
37
|
Braciak TA, Roskopf CC, Wildenhain S, Fenn NC, Schiller CB, Schubert IA, Jacob U, Honegger A, Krupka C, Subklewe M, Spiekermann K, Hopfner KP, Fey GH, Aigner M, Krause S, Mackensen A, Oduncu FS. Dual-targeting triplebody 33-16-123 (SPM-2) mediates effective redirected lysis of primary blasts from patients with a broad range of AML subtypes in combination with natural killer cells. Oncoimmunology 2018; 7:e1472195. [PMID: 30228941 PMCID: PMC6140553 DOI: 10.1080/2162402x.2018.1472195] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/13/2018] [Accepted: 04/29/2018] [Indexed: 12/20/2022] Open
Abstract
A number of agents designed for immunotherapy of Acute Myeloid Leukemia (AML) are in preclinical and early clinical development. Most of them target a single antigen on the surface of AML cells. Here we describe the development and key biological properties of a tri-specific agent, the dual-targeting triplebody SPM-2, with binding sites for target antigens CD33 and CD123, and for CD16 to engage NK cells as cytolytic effectors. Primary blasts of nearly all AML patients carry at least one of these target antigens and the pair is particularly promising for the elimination of blasts and leukemia stem cells (LSCs) from a majority of AML patients by dual-targeting agents. The cytolytic activity of NK cells mediated by SPM-2 was analyzed in vitro for primary leukemic cells from 29 patients with a broad range of AML-subtypes. Blasts from all 29 patients, including patients with genomic alterations associated with an unfavorable genetic subtype, were lysed at nanomolar concentrations of SPM-2. Maximum susceptibility was observed for cells with a combined density of CD33 and CD123 above 10,000 copies/cell. Cell populations enriched for AML-LSCs (CD34pos and CD34pos CD38neg cells) from 2 AML patients carried an increased combined antigen density and were lysed at correspondingly lower concentrations of SPM-2 than unsorted blasts. These initial findings raise the expectation that SPM-2 may also be capable of eliminating AML-LSCs and thus of prolonging survival. In the future, patients with a broad range of AML subtypes may benefit from treatment with SPM-2.
Collapse
Affiliation(s)
- Todd A. Braciak
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
| | - Claudia C. Roskopf
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
| | - Sarah Wildenhain
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nadja C. Fenn
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Christian B. Schiller
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ingo A. Schubert
- Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | - Christina Krupka
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
- Laboratory of Translational Cancer Immunol ogy, Gene Center of the LMU Munich, Munich, Germany
| | - Marion Subklewe
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
- Laboratory of Translational Cancer Immunol ogy, Gene Center of the LMU Munich, Munich, Germany
| | - Karsten Spiekermann
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
| | - Karl-Peter Hopfner
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Georg H. Fey
- Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5 - Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Krause
- Department of Internal Medicine 5 - Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Fuat S. Oduncu
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
38
|
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
|
39
|
Zhou J, Chooi JY, Ching YQ, Quah JY, Toh SHM, Ng Y, Tan TZ, Chng WJ. NF-κB promotes the stem-like properties of leukemia cells by activation of LIN28B. World J Stem Cells 2018; 10:34-42. [PMID: 29707103 PMCID: PMC5919888 DOI: 10.4252/wjsc.v10.i4.34] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/21/2018] [Accepted: 04/10/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To examine whether nuclear factor kappa B (NF-κB) activity regulates LIN28B expression and their roles in leukemia stem cell (LSC)-like properties.
METHODS We used pharmacological inhibitor and cell viability assays to examine the relation between NF-κB and LIN28B. Western blot and qRT-PCR was employed to determine their protein and mRNA levels. Luciferase reporter was constructed and applied to explore the transcriptional regulation of LIN28B. We manipulated LIN28B level in acute myeloid leukemia (AML) cells and investigated LSC-like properties with colony forming and serial replating assays.
RESULTS This study revealed the relationship between NF-κB and LIN28B in AML cells through drug inhibition and overexpression experiments. Notably, inhibition of NF-κB by pharmacological inhibitors reduced LIN28B expression and decreased cell proliferation. We demonstrated that NF-κB binds to the -819 to -811 region of LIN28B promoter, and transcriptionally regulates LIN28B expression. LIN28B protein was significantly elevated in NFκB1 transfected cells compared to vector control. Importantly, ectopic expression of LIN28B partially rescued the self-renewal capacity impaired by pharmacological inhibition of NF-κB activity.
CONCLUSION These results uncover a regulatory signaling, NF-κB/LIN28B, which plays a pivotal role in leukemia stem cell-like properties and it could serve as a promising intervening target for effective treatment of AML disease.
Collapse
Affiliation(s)
- Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Jing-Yuan Chooi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Ying Qing Ching
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Jessie Yiying Quah
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Sabrina Hui-Min Toh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Yvonne Ng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
- Department of Hematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
40
|
Latuske EM, Stamm H, Klokow M, Vohwinkel G, Muschhammer J, Bokemeyer C, Jücker M, Kebenko M, Fiedler W, Wellbrock J. Combined inhibition of GLI and FLT3 signaling leads to effective anti-leukemic effects in human acute myeloid leukemia. Oncotarget 2018; 8:29187-29201. [PMID: 28418873 PMCID: PMC5438723 DOI: 10.18632/oncotarget.16304] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 02/20/2017] [Indexed: 12/13/2022] Open
Abstract
Activation of the Hedgehog pathway has been implicated in the pathogenesis of several tumor types including myeloid leukemia. Previously we demonstrated that overexpression of Hedgehog downstream mediators GLI1/2 confers an adverse prognosis to patients with acute myeloid leukemia (AML) and is correlated with a FLT3 mutated status. To analyze a possible non-canonical activation of the Hedgehog pathway via FLT3 and PI3K, we performed blocking experiments utilizing inhibitors for FLT3 (sunitinib), PI3K (PF-04691502) and GLI1/2 (GANT61) in FLT3-mutated and FLT3 wildtype AML cell lines and primary blasts. Combination of all three compounds had stronger anti-leukemic effects in FLT3-mutated compared to FLT3 wildtype AML cells in vitro. Interestingly, the colony growth of normal CD34+ cells from healthy donors was not impeded by the triple inhibitor combination possibly opening a therapeutic window for the clinical use of inhibitor combinations. Besides, combined treatment with sunitinib, PF-04691502 and GANT61 significantly prolonged the survival of mice transplanted with FLT3-mutated MV4-11 cells compared to the single agent treatments. Furthermore, the inhibition of FLT3 and PI3K resulted in reduced GLI protein expression and promotor activity in FLT3-mutated but not in FLT3 wildtype AML cell lines in western blotting and GLI1/2 promoter assays supporting our hypothesis of non-canonical GLI activation via FLT3. In summary, FLT3-mutated in contrast to FLT3 wildtype cells or normal human hematopoietic progenitor cells are exquisitely sensitive to combined inhibition by FLT3, PI3K and GLI1/2 overcoming some of the limitations of current FLT3 directed therapy in AML. The development of GLI1/2 inhibitors is highly desirable.
Collapse
Affiliation(s)
- Emily-Marie Latuske
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hauke Stamm
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Klokow
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabi Vohwinkel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana Muschhammer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maxim Kebenko
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
41
|
Jiménez-Hernández E, Fajardo-Gutiérrez A, Núñez-Enriquez JC, Martín-Trejo JA, Espinoza-Hernández LE, Flores-Lujano J, Arellano-Galindo J, Medina-Sanson A, Paredes-Aguilera R, Merino-Pasaye LE, Velázquez-Aviña MM, Torres-Nava JR, Espinosa-Elizondo RM, Amador-Sánchez R, Dosta-Herrera JJ, Mondragón-García JA, Valdés-Guzmán H, Mejía-Pérez L, Espinoza-Anrubio G, Paz-Bribiesca MM, Salcedo-Lozada P, Landa-García RÁ, Ramírez-Colorado R, Hernández-Mora L, Pérez-Saldivar ML, Santamaría-Ascencio M, López-Loyola A, Godoy-Esquivel AH, García-López LR, Anguiano-Ávalos AI, Mora-Rico K, Castañeda-Echevarría A, Rodríguez-Jiménez R, Cibrian-Cruz JA, Solís-Labastida KA, Cárdenas-Cardos R, Martínez-Avalos A, Flores-Villegas LV, Peñaloza-González JG, González-Ávila AI, Altamirano-García MB, López-Santiago N, Sánchez-Ruiz M, Rivera-Luna R, Rodríguez-Villalobos LR, Hernández-Pérez F, Olvera-Durán JÁ, García-Cortés LR, Mata-Rocha M, Sepúlveda-Robles OA, González-Bonilla CR, Bekker-Méndez VC, Jiménez-Morales S, Rosas-Vargas H, Mejía-Aranguré JM. A greater birthweight increases the risk of acute leukemias in Mexican children-experience from the Mexican Interinstitutional Group for the Identification of the Causes of Childhood Leukemia (MIGICCL). Cancer Med 2018. [PMID: 29533016 PMCID: PMC5911591 DOI: 10.1002/cam4.1414] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In Mexico, due to the high rates of diabetes, overweight, and obesity, there has also been noted an increased newborn weight, which may be contributing to the elevated incidence rate of childhood acute leukemia (AL). We conducted a case-control study in public hospitals of Mexico City aimed to know whether a greater weight at birth is associated with a higher risk of developing leukemia. We included incident cases with acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) diagnosed between 2010 and 2015. Controls were frequency-matched to the cases by age, sex, and health institution. Logistic regression analysis was performed adjusting risks by child's sex, overcrowding index, birth order, and mother's age at the time of pregnancy. Adjusted odds ratios (aORs) and 95% confidence intervals were calculated. A total of 1455 cases and 1455 controls were included. An evident association between ALL and child's birthweight ≥2500 g was found (aOR 2.06; 95% CI: 1.59, 2.66) and also, in those with birthweight ≥3500 g (aOR 1.19; 95% CI: 1.00, 1.41). In AML patients with birthweight ≥2500 g and ≥3500 g, an aOR of 1.77 (95% CI: 1.07, 2.94) and 1.42 (95% CI: 1.03-1.95) was observed, respectively. No association was noticed with either type of AL and a birthweight ≥4000 g. To sum up, we found a moderate association between not having a low birthweight and an increased risk of acute leukemias. Birthweight ≥3500 g was also a risk factor for both types of leukemia. This suggests that a greater birthweight may increase the risk of acute leukemias in Mexican children.
Collapse
Affiliation(s)
- Elva Jiménez-Hernández
- Health Research Coordination, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico.,Pediatric Hematology Services, Hospital General "Gaudencio González Garza", CMN "La Raza", IMSS, Mexico City, Mexico
| | - Arturo Fajardo-Gutiérrez
- Medical Research Unit in Clinical Epidemiology, UMAE Hospital de Pediatría CMN "Siglo XXI", IMSS, Mexico City, Mexico
| | - Juan Carlos Núñez-Enriquez
- Medical Research Unit in Clinical Epidemiology, UMAE Hospital de Pediatría CMN "Siglo XXI", IMSS, Mexico City, Mexico
| | | | | | - Janet Flores-Lujano
- Medical Research Unit in Clinical Epidemiology, UMAE Hospital de Pediatría CMN "Siglo XXI", IMSS, Mexico City, Mexico
| | - José Arellano-Galindo
- Children's Hospital of Mexico, Federico Gómez, Secretaria de Salud (SS), Mexico City, Mexico
| | - Aurora Medina-Sanson
- Children's Hospital of Mexico, Federico Gómez, Secretaria de Salud (SS), Mexico City, Mexico
| | | | - Laura Elizabeth Merino-Pasaye
- Pediatric Hematology Services, CMN "20 de Noviembre", Instituto de Seguridad Social al Servicio de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | | | - José Refugio Torres-Nava
- Oncology Services, Hospital Pediátrico "Moctezuma", Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | | | - Raquel Amador-Sánchez
- Pediatric Hematology Service, Hospital General Regional (HGR) No. 1 "Dr. Carlos Mac Gregor Sánchez Navarro" IMSS, Mexico City, Mexico
| | - Juan José Dosta-Herrera
- Pediatric Surgery Service, Hospital General "Gaudencio González Garza", CMN "La Raza", IMSS, Mexico City, Mexico
| | | | | | | | - Gilberto Espinoza-Anrubio
- Servicio de Pediatría, Hospital General Zona (HGZ) No. 8 "Dr. Gilberto Flores Izquierdo" IMSS, Mexico City, Mexico
| | | | - Perla Salcedo-Lozada
- General Hospital of Ecatepec "Las Américas", Instituto de Salud del Estado de México (ISEM), Mexico City, Mexico
| | | | | | | | - María Luisa Pérez-Saldivar
- Medical Research Unit in Clinical Epidemiology, UMAE Hospital de Pediatría CMN "Siglo XXI", IMSS, Mexico City, Mexico
| | | | | | | | | | | | - Karina Mora-Rico
- Pediatric Surgery Service, Hospital Regional "1° Octubre", ISSSTE, Mexico City, Mexico
| | | | - Roberto Rodríguez-Jiménez
- Pediatric Services, Hospital General de Zona con Medicina Familiar (HGZMF) No. 29 IMSS, Mexico City, Mexico
| | | | | | | | | | - Luz Victoria Flores-Villegas
- Pediatric Hematology Services, CMN "20 de Noviembre", Instituto de Seguridad Social al Servicio de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | | | - Ana Itamar González-Ávila
- Pediatric Hematology Service, Hospital General Regional (HGR) No. 1 "Dr. Carlos Mac Gregor Sánchez Navarro" IMSS, Mexico City, Mexico
| | | | - Norma López-Santiago
- Hematology Services, Instituto Nacional de Pediatría (INP), SS, Mexico City, Mexico
| | - Martin Sánchez-Ruiz
- General Hospital of Ecatepec "Las Américas", Instituto de Salud del Estado de México (ISEM), Mexico City, Mexico
| | | | | | | | | | | | - Minerva Mata-Rocha
- Molecular Biology Laboratory, UMAE, Hospital de Pediatría, CMN "Siglo XXI", IMSS, Mexico City, Mexico
| | | | - Cesar Raúl González-Bonilla
- Laboratory of the Coordination of Epidemiological Surveillance and Support in Contingencies, Unidad de Investigación Médica en Inmunología e Infectología, Hospital de Infectología "Dr. Daniel Méndez Hernández", CMN "La Raza", IMSS, Mexico City, Mexico
| | - Vilma Carolina Bekker-Méndez
- Medical Research Unit in Immunology and Infectology, Hospital de Infectología "Dr. Daniel Méndez Hernández", CMN "La Raza", IMSS, Mexico City, Mexico
| | - Silvia Jiménez-Morales
- Cancer Genomics Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Haydee Rosas-Vargas
- Medical Research Unit in Human Genetics, UMAE, Hospital de Pediatría, CMN "Siglo XXI", IMSS, Mexico City, Mexico
| | - Juan Manuel Mejía-Aranguré
- Health Research Coordination, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico.,Medical Research Unit in Clinical Epidemiology, UMAE Hospital de Pediatría CMN "Siglo XXI", IMSS, Mexico City, Mexico.,Molecular Biology Laboratory, UMAE, Hospital de Pediatría, CMN "Siglo XXI", IMSS, Mexico City, Mexico
| |
Collapse
|
42
|
Forte D, Salvestrini V, Corradi G, Rossi L, Catani L, Lemoli RM, Cavo M, Curti A. The tissue inhibitor of metalloproteinases-1 (TIMP-1) promotes survival and migration of acute myeloid leukemia cells through CD63/PI3K/Akt/p21 signaling. Oncotarget 2018; 8:2261-2274. [PMID: 27903985 PMCID: PMC5356797 DOI: 10.18632/oncotarget.13664] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/21/2016] [Indexed: 12/15/2022] Open
Abstract
We and others have shown that the Tissue Inhibitor of Metalloproteinases-1 (TIMP-1), a member of the inflammatory network exerting pleiotropic effects in the bone marrow (BM) microenvironment, regulates the survival and proliferation of different cell types, including normal hematopoietic progenitor cells. Moreover, TIMP-1 has been shown to be involved in cancer progression. However, its role in leukemic microenvironment has not been addressed. Here, we investigated the activity of TIMP-1 on Acute Myelogenous Leukemia (AML) cell functions. First, we found that TIMP-1 levels were increased in the BM plasma of AML patients at diagnosis. In vitro, recombinant human (rh)TIMP-1 promoted the survival and cell cycle S-phase entry of AML cells. These kinetic effects were related to the downregulation of cyclin-dependent kinase inhibitor p21. rhTIMP-1 increases CXCL12-driven migration of leukemic cells through PI3K signaling. Interestingly, activation of CD63 receptor was required for TIMP-1's cytokine/chemokine activity. Of note, rhTIMP-1 stimulation modulated mRNA expression of Hypoxia Inducible Factor (HIF)-1α, downstream of PI3K/Akt activation. We then co-cultured AML cells with normal or leukemic mesenchymal stromal cells (MSCs) to investigate the interaction of TIMP-1 with cellular component(s) of BM microenvironment. Our results showed that the proliferation and migration of leukemic cells were greatly enhanced by rhTIMP-1 in presence of AML-MSCs as compared to normal MSCs. Thus, we demonstrated that TIMP-1 modulates leukemic blasts survival, migration and function via CD63/PI3K/Akt/p21 signaling. As a “bad actor” in a “bad soil”, we propose TIMP-1 as a potential novel therapeutic target in leukemic BM microenvironment.
Collapse
Affiliation(s)
- Dorian Forte
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Valentina Salvestrini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Giulia Corradi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Lara Rossi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Lucia Catani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Roberto M Lemoli
- Clinic of Hematology, Department of Internal Medicine (DiMI), University of Genoa, Genoa, Italy
| | - Michele Cavo
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Antonio Curti
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| |
Collapse
|
43
|
Petrov JC, Wada M, Pinz KG, Yan LE, Chen KH, Shuai X, Liu H, Chen X, Leung LH, Salman H, Hagag N, Liu F, Jiang X, Ma Y. Compound CAR T-cells as a double-pronged approach for treating acute myeloid leukemia. Leukemia 2018. [PMID: 29515236 PMCID: PMC5990523 DOI: 10.1038/s41375-018-0075-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Acute myeloid leukemia (AML) bears heterogeneous cells that can consequently offset killing by single-CAR-based therapy, which results in disease relapse. Leukemic stem cells (LSCs) associated with CD123 expression comprise a rare population that also plays an important role in disease progression and relapse. Here, we report on the robust anti-tumor activity of a compound CAR (cCAR) T-cell possessing discrete scFv domains targeting two different AML antigens, CD123, and CD33, simultaneously. We determined that the resulting cCAR T-cells possessed consistent, potent, and directed cytotoxicity against each target antigen population. Using four leukemia mouse models, we found superior in vivo survival after cCAR treatment. We also designed an alemtuzumab safety-switch that allowed for rapid cCAR therapy termination in vivo. These findings indicate that targeting both CD123 and CD33 on AML cells may be an effective strategy for eliminating both AML bulk disease and LSCs, and potentially prevent relapse due to antigen escape or LSC persistence.
Collapse
Affiliation(s)
- Jessica C Petrov
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Masayuki Wada
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA.
| | - Kevin G Pinz
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Lulu E Yan
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Kevin H Chen
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Xiao Shuai
- Department of Hematology West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hua Liu
- Department of Pathology, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | - Xi Chen
- Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Lai-Han Leung
- Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Huda Salman
- Department of Internal Medicine Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY, 11794, USA
| | - Nabil Hagag
- Department of Internal Medicine Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY, 11794, USA
| | - Fang Liu
- Department of Hematology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - Xun Jiang
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Yupo Ma
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA. .,Department of Pathology, Stony Brook Medicine, Stony Brook, NY, 11794, USA. .,Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
44
|
Nowacka JD, Baumgartner C, Pelorosso C, Roth M, Zuber J, Baccarini M. MEK1 is required for the development of NRAS-driven leukemia. Oncotarget 2018; 7:80113-80130. [PMID: 27741509 PMCID: PMC5348309 DOI: 10.18632/oncotarget.12555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/29/2016] [Indexed: 11/25/2022] Open
Abstract
The dual-specificity kinases MEK1 and MEK2 act downstream of RAS/RAF to induce ERK activation, which is generally considered protumorigenic. Activating MEK mutations have not been discovered in leukemia, in which pathway activation is caused by mutations in upstream components such as RAS or Flt3. The anti-leukemic potential of MEK inhibitors is being tested in clinical trials; however, downregulation of MEK1 promotes Eμ-Myc-driven lymphomagenesis and MEK1 ablation induces myeloproliferative disease in mice, raising the concern that MEK inhibitors may be inefficient or counterproductive in this context. We investigated the role of MEK1 in the proliferation of human leukemic cell lines and in retroviral models of leukemia. Our data show that MEK1 suppression via RNA interference and genomic engineering does not affect the proliferation of human leukemic cell lines in culture; similarly, MEK1 ablation does not impact the development of MYC-driven leukemia in vivo. In contrast, MEK1 ablation significantly reduces tumorigenesis driven by Nras alone or in combination with Myc. Thus, while MEK1 restricts proliferation and tumorigenesis in some cellular and genetic contexts, it cannot be considered a tumor suppressor in the context of leukemogenesis. On the contrary, its role in NRAS-driven leukemogenesis advocates the use of MEK inhibitors, particularly in combination with PI3K/AKT inhibitors, in hematopoietic malignancies involving RAS activation.
Collapse
Affiliation(s)
- Joanna D Nowacka
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Christian Baumgartner
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Cristiana Pelorosso
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria.,Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, A. Meyer Children's Hospital-University of Florence, Florence, Italy
| | - Mareike Roth
- Research Institute of Molecular Pathology, Vienna, Austria
| | - Johannes Zuber
- Research Institute of Molecular Pathology, Vienna, Austria
| | - Manuela Baccarini
- Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of Vienna, Vienna, Austria
| |
Collapse
|
45
|
Aziz H, Ping CY, Alias H, Ab Mutalib NS, Jamal R. Gene Mutations as Emerging Biomarkers and Therapeutic Targets for Relapsed Acute Myeloid Leukemia. Front Pharmacol 2017; 8:897. [PMID: 29270125 PMCID: PMC5725465 DOI: 10.3389/fphar.2017.00897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/24/2017] [Indexed: 12/19/2022] Open
Abstract
It is believed that there are key differences in the genomic profile between adult and childhood acute myeloid leukemia (AML). Relapse is the significant contributor of mortality in patients with AML and remains as the leading cause of cancer death among children, posing great challenges in the treatment of AML. The knowledge about the genomic lesions in childhood AML is still premature as most genomic events defined in children were derived from adult cohorts. However, the emerging technologies of next generation sequencing have narrowed the gap of knowledge in the biology of AML by the detection of gene mutations for each sub-type which have led to the improvement in terms of prognostication as well as the use of targeted therapies. In this review, we describe the recent understanding of the genomic landscape including the prevalence of mutation, prognostic impact, and targeted therapies that will provide an insight into the pathogenesis of AML relapse in both adult and childhood cases.
Collapse
Affiliation(s)
- Habsah Aziz
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Chow Y Ping
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Hamidah Alias
- Department of Paediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Rahman Jamal
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
46
|
Shi ZX, Li HY, Yang XD, Gao H, Li DG, Yang WH, Yao F, Yan LX. Yi-qi-yang-yin-tang increases the sensitivity of KG1a leukemia stem cells to daunorubicin by promoting cell cycle progression and regulating the expression of PTEN, TOPOII and mTOR. Oncol Lett 2017; 14:6441-6448. [PMID: 29163680 PMCID: PMC5686439 DOI: 10.3892/ol.2017.7067] [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: 09/09/2015] [Accepted: 07/14/2017] [Indexed: 11/29/2022] Open
Abstract
The present study aimed to investigate the effects of serum containing a combination of yi-qi-yang-yin-tang (YQYYT) and daunorubicin (DNR) on multidrug resistance in KG1a leukemia stem cells (LSCs). The effects of YQYYT and DNR on proliferation, cell cycle progression and the expression of phosphatase and tensin homolog (PTEN), topoisomerase II (Topo II) and mechanistic target of rapamycin (mTOR) in KG1a cells were investigated in vitro using cell counting kit-8 assay, flow cytometry, reverse transcription-quantitative polymerase chain reaction and western blotting, respectively. It was revealed that YQYYT-containing serum did not affect proliferation of KG1a cells compared with the blank group. Furthermore, there were no significant differences on the inhibition of proliferation among different groups at various concentrations of YQYYT. Treatment with YQYYT-containing serum (volume, 20 and 40 µl) and DNR was able to significantly inhibit the proliferation of KG1a cells compared with the blank group. The inhibition rate in the treatment group with YQYYT-containing serum (40 µl) and DNR for 48 h (72.5%) was higher compared with treatment for 24 h (60.4%, P<0.01). Treatment with YQYYT-containing serum was able to promote G0 phase of KG1a cells into cell cycle in a dose- and time-dependent manner, and significantly upregulated the mRNA expression of PTEN and Topo II, but did not affect mTOR expression compared with the blank group. Treatment with serum containing YQYYT alone did not directly affect the proliferation of KG1a cells, but when the cells were treated with a combination of YQYYT-containing serum and DNR, the proliferation of KG1a cells was significantly inhibited in a dose- and time-dependent manner. Furthermore, treatment with YQYYT-containing serum was able to promote cell cycle progression of KG1a cells in the G0 phase and upregulate the expression of the negative regulatory genes PTEN and Topo II. These results indicated the potential of YQYYT to reverse multidrug resistance in LSCs.
Collapse
Affiliation(s)
- Zhe-Xin Shi
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, P.R. China
| | - Hong-Yu Li
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Xiang-Dong Yang
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, P.R. China
| | - Hong Gao
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, P.R. China
| | - De-Guan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Academy of Medical Science and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Wen-Hua Yang
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, P.R. China
| | - Fang Yao
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, P.R. China
| | - Li-Xiang Yan
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, P.R. China
| |
Collapse
|
47
|
Parisi S, Lecciso M, Ocadlikova D, Salvestrini V, Ciciarello M, Forte D, Corradi G, Cavo M, Curti A. The More, The Better: "Do the Right Thing" For Natural Killer Immunotherapy in Acute Myeloid Leukemia. Front Immunol 2017; 8:1330. [PMID: 29097997 PMCID: PMC5653691 DOI: 10.3389/fimmu.2017.01330] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/29/2017] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells are circulating CD3− lymphocytes, which express CD56 or CD16 and an array of inhibitory receptors, called killer-immunoglobulin-like receptors (KIRs). Alloreactive KIR-ligand mismatched NK cells crucially mediate the innate immune response and have a well-recognized antitumor activity. Adoptive immunotherapy with alloreactive NK cells determined promising clinical results in terms of response in acute myeloid leukemia (AML) patients and several data demonstrated that response can be influenced by the composition of NK graft. Several data show that there is a correlation between NK alloreactivity and clinical outcome: in a cohort of AML patients who received NK infusion with active disease, more alloreactive NK cell clones were found in the donor repertoire of responders than in non-responders. These findings demonstrate that the frequency of alloreactive NK cell clones influence clinical response in AML patients undergoing NK cell immunotherapy. In this work, we will review the most recent preclinical and clinical data about the impact of alloreactive NK cells features other than frequency of alloreactive clones and cytokine network status on their anti-leukemic activity. A better knowledge of these aspects is critical to maximize the effects of this therapy in AML patients.
Collapse
Affiliation(s)
- Sarah Parisi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Mariangela Lecciso
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Darina Ocadlikova
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Valentina Salvestrini
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Marilena Ciciarello
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Dorian Forte
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Giulia Corradi
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Michele Cavo
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Antonio Curti
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology L. and A. Seràgnoli, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
48
|
Laing AA, Harrison CJ, Gibson BE, Keeshan K. Unlocking the potential of anti-CD33 therapy in adult and childhood acute myeloid leukemia. Exp Hematol 2017; 54:40-50. [DOI: 10.1016/j.exphem.2017.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 10/19/2022]
|
49
|
Paprocka M, Bielawska-Pohl A, Rossowska J, Krawczenko A, Duś D, Kiełbiński M, Haus O, Podolak-Dawidziak M, Kuliczkowski K. MRP1 protein expression in leukemic stem cells as a negative prognostic marker in acute myeloid leukemia patients. Eur J Haematol 2017; 99:415-422. [DOI: 10.1111/ejh.12938] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Maria Paprocka
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Aleksandra Bielawska-Pohl
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Agnieszka Krawczenko
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Danuta Duś
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy; The Polish Academy of Sciences; Wroclaw Poland
| | - Marek Kiełbiński
- Department and Clinic of Hematology; Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| | - Olga Haus
- Department and Clinic of Hematology; Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| | - Maria Podolak-Dawidziak
- Department and Clinic of Hematology; Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| | - Kazimierz Kuliczkowski
- Department and Clinic of Hematology; Blood Neoplasms and Bone Marrow Transplantation; Wroclaw Medical University; Wroclaw Poland
| |
Collapse
|
50
|
Key Issues Related to Cryopreservation and Storage of Stem Cells and Cancer Stem Cells: Protecting Biological Integrity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 951:1-12. [PMID: 27837550 DOI: 10.1007/978-3-319-45457-3_1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cryopreservation and biobanking of stem cells are becoming increasingly important as stem cell technology and application attract the interest of industry, academic research, healthcare and patient organisations. Stem cell are already being used in the treatment of some diseases and it is anticipated that stem cell therapy will play a central role in future medicine. Similarly, the discovery of both hematopoietic and solid tumor stem cells and their clinical relevance have profoundly altered paradigms for cancer research as the cancer stem cells are considered promising new targets against cancer. Consequently, long-term cryopreservation and banking of normal and malignant stem cells is crucial and will inevitably become a routine procedure that requires highly regulated and safe methods of specimen storage. There is, however, an increasing amount of evidence showing contradictory results on the impact of cryopreservation and thawing of stem cells, including extensive physical and biological stresses, apoptosis and necrosis, mitochondrial injuries, changes to basal respiration and ATP production, cellular structural damage, telomere shortening and cellular senescence, and DNA damage and oxidative stress. Notably, cell surface proteins that play a major role in stem cell fate and are used as the biomarkers of stem cells are more vulnerable to cold stress than other proteins. There are also data supporting the alteration in some biological features and genetic integrity at the molecular level of the post-thawed stem cells. This article reviews the current and future challenges of cryopreservation of stem cells and stresses the need for further rigorous research on the methodologies for freezing and utilizing cancer stem cells following long-term storage.
Collapse
|