283 results on '"Leukemia stem cell"'
Search Results
2. Slow‐replicating leukemia cells represent a leukemia stem cell population with high cell‐surface CD74 expression
- Author
-
Huan Li, Zhijie Cao, Yiming Liu, Zhenya Xue, Yishuang Li, Haiyan Xing, Yingxi Xu, Runxia Gu, Shaowei Qiu, Hui Wei, Min Wang, Qing Rao, and Jianxiang Wang
- Subjects
acute myeloid leukemia ,CD74 ,chemotherapy resistance ,leukemia stem cell ,quiescence ,self‐renewal ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - 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.
- Published
- 2024
- Full Text
- View/download PDF
3. Characterization of stem cell landscape and assessing the stemness degree to aid clinical therapeutics in hematologic malignancies
- Author
-
Yuan-dong Feng, Jin Du, Hong-li Chen, Ying Shen, Ya-chun Jia, Peng-yu Zhang, Aili He, and Yun Yang
- Subjects
hematologic malignancy ,Machine learning ,Leukemia stem cell ,Drug sensitivity ,Bone marrow microenvironment ,Medicine ,Science - Abstract
Abstract Hematological malignancies are a group of cancers that affect the blood, bone marrow, and lymphatic system. Cancer stem cells (CSCs) are believed to be responsible for the initiation, progression, and relapse of hematological malignancies. However, identifying and targeting CSCs presents many challenges. We aimed to develop a stemness index (HSCsi) to identify and guide the therapy targeting CSCs in hematological malignancies. We developed a novel one-class logistic regression (OCLR) algorithm to identify transcriptomic feature sets related to stemness in hematologic malignancies. We used the HSCsi to measure the stemness degree of leukemia stem cells (LSCs) and correlate it with clinical outcomes.We analyze the correlation of HSCsi with genes and pathways involved in drug resistance and immune microenvironment of acute myeloid leukemia (AML). HSCsi revealed stemness-related biological mechanisms in hematologic malignancies and effectively identify LSCs. The index also predicted survival and relapse rates of various hematologic malignancies. We also identified potential drugs and interventions targeting cancer stem cells (CSCs) of hematologic malignancies by the index. Moreover, we found a correlation between stemness and bone marrow immune microenvironment in AML. Our study provides a novel method and tool to assess the stemness degree of hematologic malignancies and its implications for clinical outcomes and therapeutic strategies. Our HSC stemness index can facilitate the precise stratification of hematologic malignancies, suggest possible targeted and immunotherapy options, and guide the selection of patients.
- Published
- 2024
- Full Text
- View/download PDF
4. Slow‐replicating leukemia cells represent a leukemia stem cell population with high cell‐surface CD74 expression.
- Author
-
Li, Huan, Cao, Zhijie, Liu, Yiming, Xue, Zhenya, Li, Yishuang, Xing, Haiyan, Xu, Yingxi, Gu, Runxia, Qiu, Shaowei, Wei, Hui, Wang, Min, Rao, Qing, and Wang, Jianxiang
- 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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Characterization of stem cell landscape and assessing the stemness degree to aid clinical therapeutics in hematologic malignancies.
- Author
-
Feng, Yuan-dong, Du, Jin, Chen, Hong-li, Shen, Ying, Jia, Ya-chun, Zhang, Peng-yu, He, Aili, and Yang, Yun
- Subjects
- *
CANCER stem cells , *HEMATOLOGIC malignancies , *ACUTE myeloid leukemia , *PATIENT selection , *LYMPHATICS - Abstract
Hematological malignancies are a group of cancers that affect the blood, bone marrow, and lymphatic system. Cancer stem cells (CSCs) are believed to be responsible for the initiation, progression, and relapse of hematological malignancies. However, identifying and targeting CSCs presents many challenges. We aimed to develop a stemness index (HSCsi) to identify and guide the therapy targeting CSCs in hematological malignancies. We developed a novel one-class logistic regression (OCLR) algorithm to identify transcriptomic feature sets related to stemness in hematologic malignancies. We used the HSCsi to measure the stemness degree of leukemia stem cells (LSCs) and correlate it with clinical outcomes.We analyze the correlation of HSCsi with genes and pathways involved in drug resistance and immune microenvironment of acute myeloid leukemia (AML). HSCsi revealed stemness-related biological mechanisms in hematologic malignancies and effectively identify LSCs. The index also predicted survival and relapse rates of various hematologic malignancies. We also identified potential drugs and interventions targeting cancer stem cells (CSCs) of hematologic malignancies by the index. Moreover, we found a correlation between stemness and bone marrow immune microenvironment in AML. Our study provides a novel method and tool to assess the stemness degree of hematologic malignancies and its implications for clinical outcomes and therapeutic strategies. Our HSC stemness index can facilitate the precise stratification of hematologic malignancies, suggest possible targeted and immunotherapy options, and guide the selection of patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. CD123表达的儿童急性淋巴细胞白血病的 临床特征及预后分析.
- Author
-
朱可夫, 李海金, 乔川芙, 王柳方, 吴佩静, 陈颖, and 田新
- Subjects
LYMPHOBLASTIC leukemia ,CHILDREN'S hospitals ,LEUKOCYTE count ,GENE fusion ,T cells - Abstract
Copyright of Chinese Journal of Contemporary Pediatrics is the property of Xiangya Medical Periodical Press and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
7. Clinical Insights into Structure, Regulation, and Targeting of ABL Kinases in Human Leukemia.
- Author
-
Wu, Andrew, Liu, Xiaohu, Fruhstorfer, Clark, and Jiang, Xiaoyan
- Subjects
- *
CHRONIC myeloid leukemia , *LEUKEMIA , *SOMATIC mutation , *HEMATOPOIETIC stem cells , *PROTEIN-tyrosine kinase inhibitors , *IMPRINTED polymers , *KINASES - Abstract
Chronic myeloid leukemia is a multistep, multi-lineage myeloproliferative disease that originates from a translocation event between chromosome 9 and chromosome 22 within the hematopoietic stem cell compartment. The resultant fusion protein BCR::ABL1 is a constitutively active tyrosine kinase that can phosphorylate multiple downstream signaling molecules to promote cellular survival and inhibit apoptosis. Currently, tyrosine kinase inhibitors (TKIs), which impair ABL1 kinase activity by preventing ATP entry, are widely used as a successful therapeutic in CML treatment. However, disease relapses and the emergence of resistant clones have become a critical issue for CML therapeutics. Two main reasons behind the persisting obstacles to treatment are the acquired mutations in the ABL1 kinase domain and the presence of quiescent CML leukemia stem cells (LSCs) in the bone marrow, both of which can confer resistance to TKI therapy. In this article, we systemically review the structural and molecular properties of the critical domains of BCR::ABL1 and how understanding the essential role of BCR::ABL1 kinase activity has provided a solid foundation for the successful development of molecularly targeted therapy in CML. Comparison of responses and resistance to multiple BCR::ABL1 TKIs in clinical studies and current combination treatment strategies are also extensively discussed in this article. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Single-cell transcriptomics reveals multiple chemoresistant properties in leukemic stem and progenitor cells in pediatric AML
- Author
-
Yongping Zhang, Shuting Jiang, Fuhong He, Yuanyuan Tian, Haiyang Hu, Li Gao, Lin Zhang, Aili Chen, Yixin Hu, Liyan Fan, Chun Yang, Bi Zhou, Dan Liu, Zihan Zhou, Yanxun Su, Lei Qin, Yi Wang, Hailong He, Jun Lu, Peifang Xiao, Shaoyan Hu, and Qian-Fei Wang
- Subjects
Residual tumor cell ,Single-cell RNA sequencing ,AML ,Chemotherapy resistance ,Leukemia stem cell ,Oxidative phosphorylation ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Cancer patients can achieve dramatic responses to chemotherapy yet retain resistant tumor cells, which ultimately results in relapse. Although xenograft model studies have identified several cellular and molecular features that are associated with chemoresistance in acute myeloid leukemia (AML), to what extent AML patients exhibit these properties remains largely unknown. Results We apply single-cell RNA sequencing to paired pre- and post-chemotherapy whole bone marrow samples obtained from 13 pediatric AML patients who had achieved disease remission, and distinguish AML clusters from normal cells based on their unique transcriptomic profiles. Approximately 50% of leukemic stem and progenitor populations actively express leukemia stem cell (LSC) and oxidative phosphorylation (OXPHOS) signatures, respectively. These clusters have a higher chance of tolerating therapy and exhibit an enhanced metabolic program in response to treatment. Interestingly, the transmembrane receptor CD69 is highly expressed in chemoresistant hematopoietic stem cell (HSC)-like populations (named the CD69 + HSC-like subpopulation). Furthermore, overexpression of CD69 results in suppression of the mTOR signaling pathway and promotion of cell quiescence and adhesion in vitro. Finally, the presence of CD69 + HSC-like cells is associated with unfavorable genetic mutations, the persistence of residual tumor cells in chemotherapy, and poor outcomes in independent pediatric and adult public AML cohorts. Conclusions Our analysis reveals leukemia stem cell and OXPHOS as two major chemoresistant features in human AML patients. CD69 may serve as a potential biomarker in defining a subpopulation of chemoresistant leukemia stem cells. These findings have important implications for targeting residual chemo-surviving AML cells.
- Published
- 2023
- Full Text
- View/download PDF
9. Cellular carcinogenesis in preleukemic conditions: drivers and defenses.
- Author
-
Koki Ueda and Kazuhiko Ikeda
- Subjects
CARCINOGENESIS ,PRELEUKEMIA ,HEMATOPOIESIS ,CATENINS ,MYELOPROLIFERATIVE neoplasms - Abstract
Acute myeloid leukemia (AML) arises from preleukemic conditions. We have investigated the pathogenesis of typical preleukemia, myeloproliferative neoplasms, and clonal hematopoiesis. Hematopoietic stem cells in both preleukemic conditions harbor recurrent driver mutations ; additional mutation provokes further malignant transformation, leading to AML onset. Although genetic alterations are defined as the main cause of malignant transformation, non-genetic factors are also involved in disease progression. In this review, we focus on a non-histone chromatin protein, high mobility group AT-hook2 (HMGA2), and a physiological p53 inhibitor, murine double minute X (MDMX). HMGA2 is mainly overexpressed by dysregulation of microRNAs or mutations in polycomb components, and provokes expansion of preleukemic clones through stem cell signature disruption. MDMX is overexpressed by altered splicing balance in myeloid malignancies. MDMX induces leukemic transformation from preleukemia via suppression of p53 and p53-independent activation of WNT/β-catenin signaling. We also discuss how these non-genetic factors can be targeted for leukemia prevention therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. The Contribution of Multiplexing Single Cell RNA Sequencing in Acute Myeloid Leukemia.
- Author
-
Madaci, Lamia, Gard, Charlyne, Nin, Sébastien, Venton, Geoffroy, Rihet, Pascal, Puthier, Denis, Loriod, Béatrice, and Costello, Régis
- Subjects
ACUTE myeloid leukemia ,RNA sequencing ,DRUG target ,CELL analysis ,PROGNOSIS - Abstract
Decades ago, the treatment for acute myeloid leukemia relied on cytarabine and anthracycline. However, advancements in medical research have introduced targeted therapies, initially employing monoclonal antibodies such as ant-CD52 and anti-CD123, and subsequently utilizing specific inhibitors that target molecular mutations like anti-IDH1, IDH2, or FLT3. The challenge lies in determining the role of these therapeutic options, considering the inherent tumor heterogeneity associated with leukemia diagnosis and the clonal drift that this type of tumor can undergo. Targeted drugs necessitate an examination of various therapeutic targets at the individual cell level rather than assessing the entire population. It is crucial to differentiate between the prognostic value and therapeutic potential of a specific molecular target, depending on whether it is found in a terminally differentiated cell with limited proliferative potential or a stem cell with robust capabilities for both proliferation and self-renewal. However, this cell-by-cell analysis is accompanied by several challenges. Firstly, the scientific aspect poses difficulties in comparing different single cell analysis experiments despite efforts to standardize the results through various techniques. Secondly, there are practical obstacles as each individual cell experiment incurs significant financial costs and consumes a substantial amount of time. A viable solution lies in the ability to process multiple samples simultaneously, which is a distinctive feature of the cell hashing technique. In this study, we demonstrate the applicability of the cell hashing technique for analyzing acute myeloid leukemia cells. By comparing it to standard single cell analysis, we establish a strong correlation in various parameters such as quality control, gene expression, and the analysis of leukemic blast markers in patients. Consequently, this technique holds the potential to become an integral part of the biological assessment of acute myeloid leukemia, contributing to the personalized and optimized management of the disease, particularly in the context of employing targeted therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Upregulation of Tim‐3 is associated with poor prognosis in acute myeloid leukemia
- Author
-
Zhengwei Wu, Jiawang Ou, Nannan Liu, Zhixiang Wang, Junjie Chen, Zihong Cai, Xiaoli Liu, Xiao Yu, Min Dai, and Hongsheng Zhou
- Subjects
acute myeloid leukemia ,immune response ,leukemia stem cell ,prognosis ,T‐cell immunoglobulin mucin 3 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy originated from leukemia stem cells (LSC). Emerging evidence suggests T‐cell immunoglobulin mucin‐3(Tim3) as surface marker for LSC. However, the clinical significance and biology of Tim‐3 in AML remain to be determined, especially those LSCs. In public AML databases as well as our data, we separated AML patients into Tim‐3high and Tim‐3low subsets using the X‐tile software and evaluated the associations between Tim‐3 and overall survival (OS) and disease‐free survival (DFS). The Cancer Genome Atlas (TCGA) cohort revealed that high Tim‐3 expression in leukemic cells was linked with poor prognosis (DFS: p = 0.018; OS: p = 0.041). Furthermore, multiple regression analysis shows that Tim‐3 was an independent factor for the prognosis (HR = 2.26, 95% CI = 1.15–4.44, p = 0.017). Validation cohort of public gene expression omnibus (GEO) confirmed that Tim‐3 was a prognostic candidate in AML. Besides, in our internal cohort, we also confirmed that over expression of Tim‐3 protein in LSC/LPC made poor prognosis in AML. Additionally, we revealed that the LSC markers AKR1C3, CD34, and MMRN1 were upregulated in the Tim‐3high group of TCGA. We found that the upregulated genes in the Tim‐3high group were mainly enriched in immune response, cytokine binding and cell adhesion molecules, and JAK–STAT signaling pathway, by gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Collectively, we revealed that, for the first time, upregulation of Tim‐3 in LSCs at the level of gene and protein expression is associated with poor prognosis and the important biological feature of Tim‐3 of LSC in AML.
- Published
- 2023
- Full Text
- View/download PDF
12. Characterization of bone marrow heterogeneity in NK-AML (M4/M5) based on single-cell RNA sequencing
- Author
-
Wenqi Wu, Zeyan Shi, Zhongyuan Tang, Huiqun Li, Xiaoke Huang, Xiaolin Liang, Jing Li, Yibin Yao, Weihua Zhao, Meiqing Wu, Jun Luo, and Zhenfang Liu
- Subjects
Single-cell RNA sequencing ,Normal karyotype acute myeloid leukemia ,Heterogeneity ,Leukemia stem cell ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Normal karyotype acute myeloid leukemia (NK-AML) is a heterogeneous hematological malignancy that contains a minor population of self-renewing leukemia stem cells (LSCs), which complicate efforts to achieve long-term survival. We performed single-cell RNA sequencing to profile 39,288 cells from 6 bone marrow (BM) aspirates including 5 NK-AML (M4/M5) patients and 1 healthy donor. The single-cell transcriptome atlas and gene expression characteristics of each cell population in NK-AML (M4/M5) and healthy BM were obtained. In addition, we identified a distinct LSC-like cluster with possible biomarkers in NK-AML (M4/M5) and verified 6 genes using qRT‒PCR and bioinformatic analyses. In conclusion, we utilized single-cell technologies to provide an atlas of NK-AML (M4/M5) cell heterogeneity, composition, and biomarkers with implications for precision medicine and targeted therapies.
- Published
- 2023
- Full Text
- View/download PDF
13. HDAC I/IIb selective inhibitor Purinostat Mesylate combined with GLS1 inhibition effectively eliminates CML stem cells
- Author
-
Qiang Qiu, Linyu yang, Yunyu Feng, Zejiang Zhu, Ning Li, Li Zheng, Yuanyuan Sun, Cong Pan, Huandi Qiu, Xue Cui, Wei He, Fang Wang, Yuyao Yi, Minghai Tang, Zhuang Yang, Yunfan Yang, Zhihui Li, Lijuan Chen, and Yiguo Hu
- Subjects
Chronic myelogenous leukemia ,Leukemia stem cell ,Selective HDAC I/IIb inhibitor ,GLS1 ,Mouse model ,Materials of engineering and construction. Mechanics of materials ,TA401-492 ,Biology (General) ,QH301-705.5 - Abstract
Purinostat Mesylate (PM) is a novel highly selective and active HDAC I/IIb inhibitor, and the injectable formulation of PM (PMF) based on the compound prescription containing cyclodextrin completely can overcome PM's poor solubility and improves its stability and pharmacokinetic properties. Here, we showed that PM effectively repressed the survival of Ph+ leukemia cells and CD34+ leukemia cells from CML patients in vitro. In vivo studies demonstrated that PMF significantly prevented BCR-ABL(T315I) induced CML progression by restraining leukemia stem cells (LSCs), which are insensitive to chemotherapy and responsible for CML relapse. Mechanism studies revealed that targeting HDAC I/IIb repressed several important factors for LSCs survival including c-Myc, β-Catenin, E2f, Ezh2, Alox5, and mTOR, as well as interrupted some critical biologic processes. Additionally, PMF increased glutamate metabolism in LSCs by increasing GLS1. The combination of PMF and glutaminase inhibitor BPTES synergistically eradicated LSCs by altering multiple key proteins and signaling pathways which are critical for LSC survival and self-renewal. Overall, our findings represent a new therapeutic strategy for eliminating LSCs by targeting HDAC I/IIb and glutaminolysis, which potentially provides a guidance for PMF clinical trials in the future for TKI resistance CML patients.
- Published
- 2023
- Full Text
- View/download PDF
14. Prospective monitoring of chronic myeloid leukemia patients from the time of TKI discontinuation: the fate of peripheral blood CD26+ leukemia stem cells.
- Author
-
Pacelli, Paola, Santoni, Adele, Sicuranza, Anna, Abruzzese, Elisabetta, Giai, Valentina, Crugnola, Monica, Annunziata, Mario, Galimberti, Sara, Iurlo, Alessandra, Luciano, Luigiana, Sorà, Federica, Fava, Carmen, Bestoso, Elena, Marzano, Cristina, Cartocci, Alessandra, Defina, Marzia, Sammartano, Vincenzo, Cencini, Emanuele, Raspadori, Donatella, and Bocchia, Monica
- Subjects
CHRONIC myeloid leukemia ,STEM cells ,LEUKEMIA ,PROTEIN-tyrosine kinase inhibitors ,DISEASE relapse - Abstract
Introduction: In chronic myeloid leukemia (CML), about half of the patients achieving a deep and stable molecular response with tyrosine kinase inhibitors (TKIs) may discontinue TKI treatment without disease recurrence. As such, treatment-free remission (TFR) has become an ambitious goal of treatment. Given the evidence that deepness and duration of molecular response are necessary but not sufficient requisites for a successful TFR, additional biological criteria are needed to identify CML patients suitable for efficacious discontinuation. Leukemia stem cells (LSCs) are supposed to be the reservoir of the disease. Previously, we demonstrated that residual circulating CD34+/CD38-/CD26+ LSCs were still detectable in a consistent number of CML patients during TFR. Methods: CML LSCs could be easily identified by flow-cytometry as they express the CD34+/CD38-/CD26+ phenotype. In this study, we explored the role of these cells and their correlation with molecular response in a cohort of 109 consecutive chronic phase CML patients prospectively monitored from the time of TKI discontinuation. Results: After a median observation time of 33 months from TKI discontinuation, 38/109 (35%) patients failed TFR after a median time of 4 months, while 71/109 (65%) patients are still in TFR. At TKI discontinuation, peripheral blood CD26+LSCs were undetectable in 48/109 (44%) patients and detectable in 61/109 (56%). No statistically significant correlation between detectable/undetectable CD26+LSCs and the rate of TFR loss was found (p = 0.616). The incidence of TFR loss based on the type of TKI treatment was statistically significant for imatinib treatment compared to that of nilotinib (p = 0.039). Exploring the behavior of CD26+LSCs during TFR, we observed fluctuating values that were very variable between patients, and they were not predictive of TFR loss. Discussion: Up to date, our results confirm that CD26+LSCs are detectable at the time of TKI discontinuation and during TFR. Moreover, at least for the observation median time of the study, the persistence of "fluctuating" values of residual CD26+LSCs does not hamper the possibility to maintain a stable TFR. On the contrary, even patients discontinuing TKI with undetectable CD26+LSCs could undergo TFR loss. Our results suggest that factors other than residual LSCs "burden" playing an active role in controlling disease recurrence. Additional studies evaluating CD26+LSCs' ability to modulate the immune system and their interaction in CML patients with very long stable TFR are ongoing. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. Prospective monitoring of chronic myeloid leukemia patients from the time of TKI discontinuation: the fate of peripheral blood CD26+ leukemia stem cells
- Author
-
Paola Pacelli, Adele Santoni, Anna Sicuranza, Elisabetta Abruzzese, Valentina Giai, Monica Crugnola, Mario Annunziata, Sara Galimberti, Alessandra Iurlo, Luigiana Luciano, Federica Sorà, Carmen Fava, Elena Bestoso, Cristina Marzano, Alessandra Cartocci, Marzia Defina, Vincenzo Sammartano, Emanuele Cencini, Donatella Raspadori, and Monica Bocchia
- Subjects
chronic myelogenous leukemia ,treatment free remission ,CD26+ ,leukemia stem cell ,TKI ,flow cytometry ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Introduction: In chronic myeloid leukemia (CML), about half of the patients achieving a deep and stable molecular response with tyrosine kinase inhibitors (TKIs) may discontinue TKI treatment without disease recurrence. As such, treatment-free remission (TFR) has become an ambitious goal of treatment. Given the evidence that deepness and duration of molecular response are necessary but not sufficient requisites for a successful TFR, additional biological criteria are needed to identify CML patients suitable for efficacious discontinuation. Leukemia stem cells (LSCs) are supposed to be the reservoir of the disease. Previously, we demonstrated that residual circulating CD34+/CD38-/CD26+ LSCs were still detectable in a consistent number of CML patients during TFR.Methods: CML LSCs could be easily identified by flow-cytometry as they express the CD34+/CD38-/CD26+ phenotype. In this study, we explored the role of these cells and their correlation with molecular response in a cohort of 109 consecutive chronic phase CML patients prospectively monitored from the time of TKI discontinuation.Results: After a median observation time of 33 months from TKI discontinuation, 38/109 (35%) patients failed TFR after a median time of 4 months, while 71/109 (65%) patients are still in TFR. At TKI discontinuation, peripheral blood CD26+LSCs were undetectable in 48/109 (44%) patients and detectable in 61/109 (56%). No statistically significant correlation between detectable/undetectable CD26+LSCs and the rate of TFR loss was found (p = 0.616). The incidence of TFR loss based on the type of TKI treatment was statistically significant for imatinib treatment compared to that of nilotinib (p = 0.039). Exploring the behavior of CD26+LSCs during TFR, we observed fluctuating values that were very variable between patients, and they were not predictive of TFR loss.Discussion: Up to date, our results confirm that CD26+LSCs are detectable at the time of TKI discontinuation and during TFR. Moreover, at least for the observation median time of the study, the persistence of “fluctuating” values of residual CD26+LSCs does not hamper the possibility to maintain a stable TFR. On the contrary, even patients discontinuing TKI with undetectable CD26+LSCs could undergo TFR loss. Our results suggest that factors other than residual LSCs “burden” playing an active role in controlling disease recurrence. Additional studies evaluating CD26+LSCs’ ability to modulate the immune system and their interaction in CML patients with very long stable TFR are ongoing.
- Published
- 2023
- Full Text
- View/download PDF
16. Upregulation of Tim‐3 is associated with poor prognosis in acute myeloid leukemia.
- Author
-
Wu, Zhengwei, Ou, Jiawang, Liu, Nannan, Wang, Zhixiang, Chen, Junjie, Cai, Zihong, Liu, Xiaoli, Yu, Xiao, Dai, Min, and Zhou, Hongsheng
- Subjects
- *
ACUTE myeloid leukemia , *HEPATITIS A virus cellular receptors , *JAK-STAT pathway , *CELL adhesion molecules , *HEMATOLOGIC malignancies , *MULTIPLE regression analysis - Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy originated from leukemia stem cells (LSC). Emerging evidence suggests T‐cell immunoglobulin mucin‐3(Tim3) as surface marker for LSC. However, the clinical significance and biology of Tim‐3 in AML remain to be determined, especially those LSCs. In public AML databases as well as our data, we separated AML patients into Tim‐3high and Tim‐3low subsets using the X‐tile software and evaluated the associations between Tim‐3 and overall survival (OS) and disease‐free survival (DFS). The Cancer Genome Atlas (TCGA) cohort revealed that high Tim‐3 expression in leukemic cells was linked with poor prognosis (DFS: p = 0.018; OS: p = 0.041). Furthermore, multiple regression analysis shows that Tim‐3 was an independent factor for the prognosis (HR = 2.26, 95% CI = 1.15–4.44, p = 0.017). Validation cohort of public gene expression omnibus (GEO) confirmed that Tim‐3 was a prognostic candidate in AML. Besides, in our internal cohort, we also confirmed that over expression of Tim‐3 protein in LSC/LPC made poor prognosis in AML. Additionally, we revealed that the LSC markers AKR1C3, CD34, and MMRN1 were upregulated in the Tim‐3high group of TCGA. We found that the upregulated genes in the Tim‐3high group were mainly enriched in immune response, cytokine binding and cell adhesion molecules, and JAK–STAT signaling pathway, by gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Collectively, we revealed that, for the first time, upregulation of Tim‐3 in LSCs at the level of gene and protein expression is associated with poor prognosis and the important biological feature of Tim‐3 of LSC in AML. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Characterization of bone marrow heterogeneity in NK-AML (M4/M5) based on single-cell RNA sequencing.
- Author
-
Wu, Wenqi, Shi, Zeyan, Tang, Zhongyuan, Li, Huiqun, Huang, Xiaoke, Liang, Xiaolin, Li, Jing, Yao, Yibin, Zhao, Weihua, Wu, Meiqing, Luo, Jun, and Liu, Zhenfang
- Subjects
- *
RNA sequencing , *BONE marrow , *ACUTE myeloid leukemia , *HETEROGENEITY , *HEMATOLOGIC malignancies - Abstract
Normal karyotype acute myeloid leukemia (NK-AML) is a heterogeneous hematological malignancy that contains a minor population of self-renewing leukemia stem cells (LSCs), which complicate efforts to achieve long-term survival. We performed single-cell RNA sequencing to profile 39,288 cells from 6 bone marrow (BM) aspirates including 5 NK-AML (M4/M5) patients and 1 healthy donor. The single-cell transcriptome atlas and gene expression characteristics of each cell population in NK-AML (M4/M5) and healthy BM were obtained. In addition, we identified a distinct LSC-like cluster with possible biomarkers in NK-AML (M4/M5) and verified 6 genes using qRT‒PCR and bioinformatic analyses. In conclusion, we utilized single-cell technologies to provide an atlas of NK-AML (M4/M5) cell heterogeneity, composition, and biomarkers with implications for precision medicine and targeted therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. CDK7/12/13 inhibition targets an oscillating leukemia stem cell network and synergizes with venetoclax in acute myeloid leukemia
- Author
-
Lixiazi He, Christian Arnold, Judith Thoma, Christian Rohde, Maksim Kholmatov, Swati Garg, Cheng‐Chih Hsiao, Linda Viol, Kaiqing Zhang, Rui Sun, Christina Schmidt, Maike Janssen, Tara MacRae, Karin Huber, Christian Thiede, Josée Hébert, Guy Sauvageau, Julia Spratte, Herbert Fluhr, Gabriela Aust, Carsten Müller‐Tidow, Christof Niehrs, Gislene Pereira, Jörg Hamann, Motomu Tanaka, Judith B Zaugg, and Caroline Pabst
- Subjects
AML ,CDK7 inhibition ,GPR56 ,leukemia stem cell ,self‐renewal ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract The heterogeneous response of acute myeloid leukemia (AML) to current anti‐leukemic therapies is only partially explained by mutational heterogeneity. We previously identified GPR56 as a surface marker associated with poor outcome across genetic groups, which characterizes two leukemia stem cell (LSC)‐enriched compartments with different self‐renewal capacities. How these compartments self‐renew remained unclear. Here, we show that GPR56+ LSC compartments are promoted in a complex network involving epithelial‐to‐mesenchymal transition (EMT) regulators besides Rho, Wnt, and Hedgehog (Hh) signaling. Unexpectedly, Wnt pathway inhibition increased the more immature, slowly cycling GPR56+CD34+ fraction and Hh/EMT gene expression, while Wnt activation caused opposite effects. Our data suggest that the crucial role of GPR56 lies in its ability to co‐activate these opposing signals, thus ensuring the constant supply of both LSC subsets. We show that CDK7 inhibitors suppress both LSC‐enriched subsets in vivo and synergize with the Bcl‐2 inhibitor venetoclax. Our data establish reciprocal transition between LSC compartments as a novel concept underlying the poor outcome in GPR56high AML and propose combined CDK7 and Bcl‐2 inhibition as LSC‐directed therapy in this disease.
- Published
- 2022
- Full Text
- View/download PDF
19. Preclinical Evaluation of the Multiple Tyrosine Kinases Inhibitor Anlotinib in Leukemia Stem Cells.
- Author
-
Jiang, Yuelong, Liu, Long, Jiang, Yirong, Li, Zhifeng, Feng, Liying, Zhuang, Xinguo, Lin, Zhijuan, Chen, Qiuling, Chen, Guoshu, He, Jixiang, Li, Guowei, Zha, Jie, and Xu, Bing
- Subjects
- *
STEM cells , *LEUKEMIA , *BCL-2 proteins , *ACUTE myeloid leukemia , *PROTEIN-tyrosine kinase inhibitors , *PROGRAMMED cell death 1 receptors , *KINASES , *ANLOTINIB - Abstract
Leukemia stem cells (LSCs) constitute the critical barrier to the cure of acute myeloid leukemia (AML) due to their chemoresistance and immune evasion property. Herein, the role of anlotinib, a multiple tyrosine kinase inhibitor, in killing LSCs and regulating chemoresistance and immune evasion was explored. Anlotinib treatment induced apoptosis of LSC-like cells as well as primary AML LSCs, while sparing the normal mononuclear cells in vitro. Moreover, anlotinib could impair the regeneration capacity of LSCs in the patient-derived leukemia xenograft mouse model. Mechanistically, anlotinib inhibited phosphorylation of c-kit, JAK2/STAT3, and STAT5, and downregulated STAT3 and STAT5 expression. In addition, anlotinib downregulated the anti-apoptotic proteins Bcl-2 and Bcl-xL, and upregulated Bax, thereby enhancing the sensitivity of LSCs to idarubicin in vitro. Intriguingly, anlotinib could also partially rescue the interferon-g production of T cells cocultured with LSCs by downregulating PD-L1 expression. In conclusion, anlotinib showed anti-LSC activity and the potential to enhance the sensitivity to idarubicin and inhibit the immunosuppressive feature of LSCs via JAK2/STAT signaling pathway downregulation in the preclinical study. Our results provided a rational basis for combinatory strategies involving anlotinib and chemotherapy or immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. Defective ribosome assembly impairs leukemia progression in a murine model of acute myeloid leukemia.
- Author
-
Sjövall D, Ghosh S, Fernandez-Fuentes N, Velasco-Hernandez T, Hogmalm A, Menendez P, Hansson J, Guibentif C, and Jaako P
- Subjects
- Animals, Mice, Humans, Tumor Suppressor Protein p53 metabolism, Mice, Inbred C57BL, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute genetics, Ribosomes metabolism, Disease Models, Animal, Disease Progression
- Abstract
Despite an advanced understanding of disease mechanisms, the current therapeutic regimen fails to cure most patients with acute myeloid leukemia (AML). In the present study, we address the role of ribosome assembly in leukemia cell function. We apply patient datasets and murine models to demonstrate that immature leukemia cells in mixed-lineage leukemia-rearranged AML are characterized by relatively high ribosome biogenesis and protein synthesis rates. Using a model with inducible regulation of ribosomal subunit joining, we show that defective ribosome assembly extends survival in mice with AML. Single-cell RNA sequencing and proteomic analyses reveal that leukemia cell adaptation to defective ribosome assembly is associated with an increase in ribosome biogenesis and deregulation of the transcription factor landscape. Finally, we demonstrate that defective ribosome assembly shows antileukemia efficacy in p53-deficient AML. Our study unveils the critical requirement of a high protein synthesis rate for leukemia progression and highlights ribosome assembly as a therapeutic target in AML., Competing Interests: Declaration of interests P.M. is co-founder of OneChain Immunotherapeutics, a spin-off company from the Josep Carreras Leukemia Research Institute with scientific interests unrelated to this work., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. IL-21/IL-21R signaling renders acute myeloid leukemia stem cells more susceptible to cytarabine treatment and CAR T cell therapy.
- Author
-
Rubino V, Hüppi M, Höpner S, Tortola L, Schnüriger N, Legenne H, Taylor L, Voggensperger S, Keller I, Bruggman R, Kronig MN, Bacher U, Kopf M, Ochsenbein AF, and Riether C
- Subjects
- Animals, Humans, Mice, Immunotherapy, Adoptive methods, Female, Mice, Inbred C57BL, Male, Receptors, Interleukin-21 metabolism, Receptors, Interleukin-21 genetics, Cell Differentiation drug effects, Xenograft Model Antitumor Assays, Cell Line, Tumor, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes drug effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute metabolism, Cytarabine pharmacology, Cytarabine therapeutic use, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Interleukins metabolism, Signal Transduction drug effects
- Abstract
Self-renewal programs in leukemia stem cells (LSCs) predict poor prognosis in patients with acute myeloid leukemia (AML). We identify CD4
+ T cell-derived interleukin (IL)-21 as an important negative regulator of self-renewal of LSCs. IL-21/IL-21R signaling favors asymmetric cell division and differentiation in LSCs through the activation of p38-MAPK signaling, resulting in reduced LSC numbers and significantly prolonged survival in murine AML models. In human AML, serum IL-21 at diagnosis is identified as an independent positive prognostic biomarker for outcome and correlates with improved survival and higher complete remission rates in patients that underwent high-dose chemotherapy. IL-21 treatment inhibits primary LSC function and enhances the effect of cytarabine and CD70 CAR T cell treatment on LSCs in vitro. Low-dose IL-21 treatment prolongs the survival of AML mice in syngeneic and xenograft experiments. Therefore, promoting IL-21/IL-21R signaling on LSCs may be an approach to reduce stemness and increase differentiation in AML., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
22. Non-Coding RNAs Are Implicit in Chronic Myeloid Leukemia Therapy Resistance.
- Author
-
Rudich, Alexander, Garzon, Ramiro, and Dorrance, Adrienne
- Subjects
- *
CHRONIC myeloid leukemia , *CIRCULAR RNA , *NON-coding RNA , *LINCRNA , *MYELOPROLIFERATIVE neoplasms , *PROTEIN-tyrosine kinase inhibitors - Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm initiated by the presence of the fusion gene BCR::ABL1. The development of tyrosine kinase inhibitors (TKIs) highly specific to p210BCR-ABL1, the constitutively active tyrosine kinase encoded by BCR::ABL1, has greatly improved the prognosis for CML patients. Now, the survival rate of CML nearly parallels that of age matched controls. However, therapy resistance remains a persistent problem in the pursuit of a cure. TKI resistance can be attributed to both BCR::ABL1 dependent and independent mechanisms. Recently, the role of non-coding RNAs (ncRNAs) has been increasingly explored due to their frequent dysregulation in a variety of malignancies. Specifically, microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs) have been shown to contribute to the development and progression of therapy resistance in CML. Since each ncRNA exhibits multiple functions and is capable of controlling gene expression, they exert their effect on CML resistance through a diverse set of mechanisms and pathways. In most cases ncRNAs with tumor suppressing functions are silenced in CML, while those with oncogenic properties are overexpressed. Here, we discuss the relevance of many aberrantly expressed ncRNAs and their effect on therapy resistance in CML. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
23. Blast cells surviving acute myeloid leukemia induction therapy are in cycle with a signature of FOXM1 activity
- Author
-
Mark S. Williams, Naseer J. Basma, Fabio M. R. Amaral, Daniel H. Wiseman, and Tim C. P. Somervaille
- Subjects
Acute myeloid leukemia ,Drug resistance ,FOXM1 ,Leukemia stem cell ,Transcriptome ,Quiescence ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Disease relapse remains common following treatment of acute myeloid leukemia (AML) and is due to chemoresistance of leukemia cells with disease repopulating potential. To date, attempts to define the characteristics of in vivo resistant blasts have focused on comparisons between leukemic cells at presentation and relapse. However, further treatment responses are often seen following relapse, suggesting that most blasts remain chemosensitive. We sought to characterise in vivo chemoresistant blasts by studying the transcriptional and genetic features of blasts from before and shortly after induction chemotherapy using paired samples from six patients with primary refractory AML. Methods Leukemic blasts were isolated by fluorescence-activated cell sorting. Fluorescence in situ hybridization (FISH), targeted genetic sequencing and detailed immunophenotypic analysis were used to confirm that sorted cells were leukemic. Sorted blasts were subjected to RNA sequencing. Lentiviral vectors expressing short hairpin RNAs were used to assess the effect of FOXM1 knockdown on colony forming capacity, proliferative capacity and apoptosis in cell lines, primary AML cells and CD34+ cells from healthy donors. Results Molecular genetic analysis revealed early clonal selection occurring after induction chemotherapy. Immunophenotypic characterisation found leukemia-associated immunophenotypes in all cases that persisted following treatment. Despite the genetic heterogeneity of the leukemias studied, transcriptional analysis found concerted changes in gene expression in resistant blasts. Remarkably, the gene expression signature suggested that post-chemotherapy blasts were more proliferative than those at presentation. Resistant blasts also appeared less differentiated and expressed leukemia stem cell (LSC) maintenance genes. However, the proportion of immunophenotypically defined LSCs appeared to decrease following treatment, with implications for the targeting of these cells on the basis of cell surface antigen expression. The refractory gene signature was highly enriched with targets of the transcription factor FOXM1. shRNA knockdown experiments demonstrated that the viability of primary AML cells, but not normal CD34+ cells, depended on FOXM1 expression. Conclusions We found that chemorefractory blasts from leukemias with varied genetic backgrounds expressed a common transcriptional program. In contrast to the notion that LSC quiescence confers resistance to chemotherapy we find that refractory blasts are both actively proliferating and enriched with LSC maintenance genes. Using primary patient material from a relevant clinical context we also provide further support for the role of FOXM1 in chemotherapy resistance, proliferation and stem cell function in AML.
- Published
- 2021
- Full Text
- View/download PDF
24. Editorial: Characteristics and behaviors of hematopoietic stem cells and leukemia stem cells in hematologic malignancies
- Author
-
Haojian Zhang and Pengxu Qian
- Subjects
leukemia stem cell ,hematopoietic stem cell ,hematopoietic malignancies ,chronic myeloid leukemia ,acute myeloid leukemia ,drug resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2022
- Full Text
- View/download PDF
25. Leukemia stem cell-bone marrow microenvironment interplay in acute myeloid leukemia development
- Author
-
Yiyi Yao, Fenglin Li, Jiansong Huang, Jie Jin, and Huafeng Wang
- Subjects
Acute myeloid leukemia ,Bone marrow microenvironment ,Leukemia stem cell ,Interaction ,Environment-mediated drug resistance ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Despite the advances in intensive chemotherapy regimens and targeted therapies, overall survival (OS) of acute myeloid leukemia (AML) remains unfavorable due to inevitable chemotherapy resistance and high relapse rate, which mainly caused by the persistence existence of leukemia stem cells (LSCs). Bone marrow microenvironment (BMM), the home of hematopoiesis, has been considered to play a crucial role in both hematopoiesis and leukemogenesis. When interrupted by the AML cells, a malignant BMM formed and thus provided a refuge for LSCs and protecting them from the cytotoxic effects of chemotherapy. In this review, we summarized the alterations in the bidirectional interplay between hematopoietic cells and BMM in the normal/AML hematopoietic environment, and pointed out the key role of these alterations in pathogenesis and chemotherapy resistance of AML. Finally, we focused on the current potential BMM-targeted strategies together with future prospects and challenges. Accordingly, while further research is necessary to elucidate the underlying mechanisms behind LSC–BMM interaction, targeting the interaction is perceived as a potential therapeutic strategy to eradicate LSCs and ultimately improve the outcome of AML.
- Published
- 2021
- Full Text
- View/download PDF
26. Leukemic Stem Cell: A Mini-Review on Clinical Perspectives.
- Author
-
Barreto, Igor Valentim, Pessoa, Flávia Melo Cunha de Pinho, Machado, Caio Bezerra, Pantoja, Laudreísa da Costa, Ribeiro, Rodrigo Monteiro, Lopes, Germison Silva, Amaral de Moraes, Maria Elisabete, de Moraes Filho, Manoel Odorico, de Souza, Lucas Eduardo Botelho, Burbano, Rommel Mário Rodriguez, Khayat, André Salim, and Moreira-Nunes, Caroline Aquino
- Subjects
STEM cells ,HEMATOPOIETIC stem cells ,HEMATOPOIETIC system ,DRUG resistance ,BONE marrow - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
27. CNST is Characteristic of Leukemia Stem Cells and is Associated With Poor Prognosis in AML.
- Author
-
Liu, Haoyu, Zhang, Xu, Zhao, Ziyan, Zhu, Hongying, Li, Danyang, Yang, Yang, Zhao, Wenbo, Zhang, Fei, Wang, Yuefeng, Zhu, Lina, Ding, Zewen, and Li, Xiangzhi
- Subjects
STEM cells ,ACUTE myeloid leukemia ,LEUKEMIA ,HEMATOPOIETIC system ,PROGNOSIS ,SURVIVAL analysis (Biometry) ,BREAST cancer prognosis ,CYTOPLASMIC granules - Abstract
Consortin (CNST) is a protein located on the trans-Golgi network that can target transmembrane proteins to the plasma membrane. Although CNST was discovered more than 10 years ago, there are still not enough studies on its function. During our search for possible new acute myeloid leukemia (AML) markers, we found that CNST was overexpressed in almost all patients with AML. By analyzing profiling data from public databases, we found that CNST expression inversely correlated with overall survival among AML patients. There was a great variation in CNST expression among different subtypes of AML, and the expression was the highest in the t(8,21) subtype, which was probably due to the direct regulation of CNST transcription by RUNX1-RUNX1T1. In addition, we analyzed the expression of CNST in different cells of the hematopoietic system. We found that CNST was associated with the low differentiation degrees of hematopoietic cells and had the highest expression level in leukemia stem cells (LSCs). Finally, we analyzed the CNST-related gene network and found that the genes negatively correlated with CNST are involved in various immune-related pathways, which indicates that CNST is likely related to immune evasion, LSC niche retention, and assembly of stress granules. In conclusion, our study suggests that CNST has the potential to be a diagnostic and prognostic biomarker for AML. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
28. Cancer non-stem cells as a potent regulator of tumor microenvironment: a lesson from chronic myeloid leukemia
- Author
-
Naofumi Mukaida, Yamato Tanabe, and Tomohisa Baba
- Subjects
Basophil ,Cancer stem cell ,Leukemia stem cell ,Megakaryocyte ,Tissue-resident stem cell ,Medicine - Abstract
Abstract A limited subset of human leukemia cells has a self-renewal capacity and can propagate leukemia upon their transplantation into animals, and therefore, are named as leukemia stem cells, in the early 1990’s. Subsequently, cell subpopulations with similar characteristics were detected in various kinds of solid cancers and were denoted as cancer stem cells. Cancer stem cells are presently presumed to be crucially involved in malignant progression of solid cancer: chemoresitance, radioresistance, immune evasion, and metastasis. On the contrary, less attention has been paid to cancer non-stem cell population, which comprise most cancer cells in cancer tissues, due to the lack of suitable markers to discriminate cancer non-stem cells from cancer stem cells. Chronic myeloid leukemia stem cells generate a larger number of morphologically distinct non-stem cells. Moreover, accumulating evidence indicates that poor prognosis is associated with the increases in these non-stem cells including basophils and megakaryocytes. We will discuss the potential roles of cancer non-stem cells in fostering tumor microenvironment, by illustrating the roles of chronic myeloid leukemia non-stem cells including basophils and megakaryocytes in the pathogenesis of chronic myeloid leukemia, a typical malignant disorder arising from leukemic stem cells.
- Published
- 2021
- Full Text
- View/download PDF
29. Leukemic Stem Cell: A Mini-Review on Clinical Perspectives
- Author
-
Igor Valentim Barreto, Flávia Melo Cunha de Pinho Pessoa, Caio Bezerra Machado, Laudreísa da Costa Pantoja, Rodrigo Monteiro Ribeiro, Germison Silva Lopes, Maria Elisabete Amaral de Moraes, Manoel Odorico de Moraes Filho, Lucas Eduardo Botelho de Souza, Rommel Mário Rodriguez Burbano, André Salim Khayat, and Caroline Aquino Moreira-Nunes
- Subjects
hematopoietic stem cells ,leukemia stem cell ,molecular biomarkers ,clinical relapse ,drug resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - 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.
- Published
- 2022
- Full Text
- View/download PDF
30. CNST is Characteristic of Leukemia Stem Cells and is Associated With Poor Prognosis in AML
- Author
-
Haoyu Liu, Xu Zhang, Ziyan Zhao, Hongying Zhu, Danyang Li, Yang Yang, Wenbo Zhao, Fei Zhang, Yuefeng Wang, Lina Zhu, Zewen Ding, and Xiangzhi Li
- Subjects
acute myeloid leukemia ,consortin ,leukemia stem cell ,prognosis ,biomarker ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Consortin (CNST) is a protein located on the trans-Golgi network that can target transmembrane proteins to the plasma membrane. Although CNST was discovered more than 10 years ago, there are still not enough studies on its function. During our search for possible new acute myeloid leukemia (AML) markers, we found that CNST was overexpressed in almost all patients with AML. By analyzing profiling data from public databases, we found that CNST expression inversely correlated with overall survival among AML patients. There was a great variation in CNST expression among different subtypes of AML, and the expression was the highest in the t(8,21) subtype, which was probably due to the direct regulation of CNST transcription by RUNX1-RUNX1T1. In addition, we analyzed the expression of CNST in different cells of the hematopoietic system. We found that CNST was associated with the low differentiation degrees of hematopoietic cells and had the highest expression level in leukemia stem cells (LSCs). Finally, we analyzed the CNST-related gene network and found that the genes negatively correlated with CNST are involved in various immune-related pathways, which indicates that CNST is likely related to immune evasion, LSC niche retention, and assembly of stress granules. In conclusion, our study suggests that CNST has the potential to be a diagnostic and prognostic biomarker for AML.
- Published
- 2022
- Full Text
- View/download PDF
31. Single-cell transcriptomics reveals multiple chemoresistant properties in leukemic stem and progenitor cells in pediatric AML
- Author
-
Zhang, Yongping, Jiang, Shuting, He, Fuhong, Tian, Yuanyuan, Hu, Haiyang, Gao, Li, Zhang, Lin, Chen, Aili, Hu, Yixin, Fan, Liyan, Yang, Chun, Zhou, Bi, Liu, Dan, Zhou, Zihan, Su, Yanxun, Qin, Lei, Wang, Yi, He, Hailong, Lu, Jun, Xiao, Peifang, Hu, Shaoyan, and Wang, Qian-Fei
- Published
- 2023
- Full Text
- View/download PDF
32. CDK7/12/13 inhibition targets an oscillating leukemia stem cell network and synergizes with venetoclax in acute myeloid leukemia.
- Author
-
He, Lixiazi, Arnold, Christian, Thoma, Judith, Rohde, Christian, Kholmatov, Maksim, Garg, Swati, Hsiao, Cheng‐Chih, Viol, Linda, Zhang, Kaiqing, Sun, Rui, Schmidt, Christina, Janssen, Maike, MacRae, Tara, Huber, Karin, Thiede, Christian, Hébert, Josée, Sauvageau, Guy, Spratte, Julia, Fluhr, Herbert, and Aust, Gabriela
- Abstract
The heterogeneous response of acute myeloid leukemia (AML) to current anti‐leukemic therapies is only partially explained by mutational heterogeneity. We previously identified GPR56 as a surface marker associated with poor outcome across genetic groups, which characterizes two leukemia stem cell (LSC)‐enriched compartments with different self‐renewal capacities. How these compartments self‐renew remained unclear. Here, we show that GPR56+ LSC compartments are promoted in a complex network involving epithelial‐to‐mesenchymal transition (EMT) regulators besides Rho, Wnt, and Hedgehog (Hh) signaling. Unexpectedly, Wnt pathway inhibition increased the more immature, slowly cycling GPR56+CD34+ fraction and Hh/EMT gene expression, while Wnt activation caused opposite effects. Our data suggest that the crucial role of GPR56 lies in its ability to co‐activate these opposing signals, thus ensuring the constant supply of both LSC subsets. We show that CDK7 inhibitors suppress both LSC‐enriched subsets in vivo and synergize with the Bcl‐2 inhibitor venetoclax. Our data establish reciprocal transition between LSC compartments as a novel concept underlying the poor outcome in GPR56high AML and propose combined CDK7 and Bcl‐2 inhibition as LSC‐directed therapy in this disease. Synopsis: RNA‐ and ATAC‐seq profiling combined with functional in vitro and in vivo studies unravel the multi‐faceted roles of GPR56, a surface marker associated with high leukemia stem cell (LSC) burden and poor prognosis in acute myeloid leukemia (AML). ATAC‐seq profiling of 35 primary AML specimens links high GPR56 expression to Wnt and Hh signaling.GPR56 is required for in vitro and in vivo expansion of primary human AML cells.GPR56 enhances besides RhoA also TGFB, Hedgehog, and Wnt pathway activities, which inhibit each other to coordinate reciprocal transition between the GPR56+CD34+ and GPR56+CD34− compartments to sustain the LSC pool.CDK7 inhibitors synergize with the Bcl‐2 inhibitor venetoclax to suppress both GPR56+ LSC‐enriched compartments in vitro and in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
33. WT1 facilitates the self-renewal of leukemia-initiating cells through the upregulation of BCL2L2: WT1-BCL2L2 axis as a new acute myeloid leukemia therapy target
- Author
-
Bin Zhou, Xianghong Jin, Weiwei Jin, Xingzhou Huang, Yanfei Wu, Haiying Li, Weijian Zhu, Xiaoyi Qin, Haige Ye, and Shenmeng Gao
- Subjects
Leukemia-initiating cell ,Leukemia stem cell ,Deubiquitinase inhibitor ,Ubiquitin–proteasome signal ,Wilms’ tumor-1 ,Self-renewal ,Medicine - Abstract
Abstract Background Overexpression of Wilms’ tumor-1 (WT1) transcription factor facilitates proliferation in acute myeloid leukemia (AML). However, whether WT1 is enriched in the leukemia-initiating cells (LICs) and leukemia stem cells (LSCs) and facilitates the self-renewal of LSCs remains poorly understood. Methods MLL-AF9-induced murine leukemia model was used to evaluate the effect of knockdown of wt1 on the self-renewal ability of LSC. RNA sequencing was performed on WT1-overexpressing cells to select WT1 targets. Apoptosis and colony formation assays were used to assess the anti-leukemic potential of a deubiquitinase inhibitor WP1130. Furthermore, NOD/SCID-IL2Rγ (NSG) AML xenotransplantation and MLL-AF9-induced murine leukemia models were used to evaluate the anti-leukemogenic potential of WP1130 in vivo. Results We found that wt1 is highly expressed in LICs and LSCs and facilitates the maintenance of leukemia in a murine MLL-AF9-induced model of AML. WT1 enhanced the self-renewal of LSC by increasing the expression of BCL2L2, a member of B cell lymphoma 2 (BCL2) family, by direct binding to its promoter region. Loss of WT1 impaired self-renewal ability in LSC and delayed the progression of leukemia. WP1130 was found to modify the WT1-BCL2L2 axis, and WP1130-induced anti-leukemic activity was mediated by ubiquitin proteasome-mediated destruction of WT1 protein. WP1130 induced apoptosis and decreased colony formation abilities of leukemia cells and prolonged the overall survival in the THP1-based xenograft NSG mouse model. WP1130 also decreased the frequency of LSC and prolonged the overall survival in MLL-AF9-induced murine leukemia model. Mechanistically, WP1130 induced the degradation of WT1 by positively affecting the ubiquitination of WT1 protein. Conclusions Our results indicate that WT1 is required for the development of AML. WP1130 exhibits anti-leukemic activity by inhibiting the WT1-BCL2L2 axis, which may represent a new acute myeloid leukemia therapy target.
- Published
- 2020
- Full Text
- View/download PDF
34. Mitochondrial Effects on Seeds of Cancer Survival in Leukemia
- Author
-
Hend E. El-Shaqanqery, Rania Hassan Mohamed, and Ahmed A. Sayed
- Subjects
leukemia ,leukemia stem cell ,metabolism ,mitochondria ,mitophagy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The cancer metabolic alteration is considered a hallmark and fast becoming a road for therapeutic intervention. Mitochondria have been regarded as essential cell elements that fuel the metabolic needs of most cancer cell types. Leukemia stem cells (LSCs) are a heterogeneous, highly self-renewing, and pluripotent cell population within leukemic cells. The most important source of ATP and metabolites to fulfill the bioenergetics and biosynthetic needs of most cancer stem cells is the mitochondria. In addition, mitochondria have a core role in autophagy and cell death and are the main source of reactive oxygen species (ROS) generation. Overall, growing evidence now shows that mitochondrial activities and pathways have changed to adapt with different types of leukemia, thus mitochondrial metabolism could be targeted for blood malignancy therapy. This review focuses on the function of mitochondria in LSC of the different leukemia types.
- Published
- 2021
- Full Text
- View/download PDF
35. Blast cells surviving acute myeloid leukemia induction therapy are in cycle with a signature of FOXM1 activity.
- Author
-
Williams, Mark S., Basma, Naseer J., Amaral, Fabio M. R., Wiseman, Daniel H., and Somervaille, Tim C. P.
- Subjects
- *
ACUTE myeloid leukemia , *CELL surface antigens , *FLUORESCENCE in situ hybridization , *RNA sequencing , *CELL physiology , *RNA metabolism , *FLOW cytometry , *CELL differentiation , *CHRONIC myeloid leukemia , *COLONY-forming units assay , *APOPTOSIS , *DISEASE relapse , *IMMUNOPHENOTYPING , *STEM cells , *GENES , *RESEARCH funding , *DRUG resistance in cancer cells - Abstract
Background: Disease relapse remains common following treatment of acute myeloid leukemia (AML) and is due to chemoresistance of leukemia cells with disease repopulating potential. To date, attempts to define the characteristics of in vivo resistant blasts have focused on comparisons between leukemic cells at presentation and relapse. However, further treatment responses are often seen following relapse, suggesting that most blasts remain chemosensitive. We sought to characterise in vivo chemoresistant blasts by studying the transcriptional and genetic features of blasts from before and shortly after induction chemotherapy using paired samples from six patients with primary refractory AML.Methods: Leukemic blasts were isolated by fluorescence-activated cell sorting. Fluorescence in situ hybridization (FISH), targeted genetic sequencing and detailed immunophenotypic analysis were used to confirm that sorted cells were leukemic. Sorted blasts were subjected to RNA sequencing. Lentiviral vectors expressing short hairpin RNAs were used to assess the effect of FOXM1 knockdown on colony forming capacity, proliferative capacity and apoptosis in cell lines, primary AML cells and CD34+ cells from healthy donors.Results: Molecular genetic analysis revealed early clonal selection occurring after induction chemotherapy. Immunophenotypic characterisation found leukemia-associated immunophenotypes in all cases that persisted following treatment. Despite the genetic heterogeneity of the leukemias studied, transcriptional analysis found concerted changes in gene expression in resistant blasts. Remarkably, the gene expression signature suggested that post-chemotherapy blasts were more proliferative than those at presentation. Resistant blasts also appeared less differentiated and expressed leukemia stem cell (LSC) maintenance genes. However, the proportion of immunophenotypically defined LSCs appeared to decrease following treatment, with implications for the targeting of these cells on the basis of cell surface antigen expression. The refractory gene signature was highly enriched with targets of the transcription factor FOXM1. shRNA knockdown experiments demonstrated that the viability of primary AML cells, but not normal CD34+ cells, depended on FOXM1 expression.Conclusions: We found that chemorefractory blasts from leukemias with varied genetic backgrounds expressed a common transcriptional program. In contrast to the notion that LSC quiescence confers resistance to chemotherapy we find that refractory blasts are both actively proliferating and enriched with LSC maintenance genes. Using primary patient material from a relevant clinical context we also provide further support for the role of FOXM1 in chemotherapy resistance, proliferation and stem cell function in AML. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
36. Mitochondrial Effects on Seeds of Cancer Survival in Leukemia.
- Author
-
El-Shaqanqery, Hend E., Mohamed, Rania Hassan, and Sayed, Ahmed A.
- Subjects
CANCER stem cells ,LEUKEMIA ,CELL populations ,MITOCHONDRIA ,REACTIVE oxygen species - Abstract
The cancer metabolic alteration is considered a hallmark and fast becoming a road for therapeutic intervention. Mitochondria have been regarded as essential cell elements that fuel the metabolic needs of most cancer cell types. Leukemia stem cells (LSCs) are a heterogeneous, highly self-renewing, and pluripotent cell population within leukemic cells. The most important source of ATP and metabolites to fulfill the bioenergetics and biosynthetic needs of most cancer stem cells is the mitochondria. In addition, mitochondria have a core role in autophagy and cell death and are the main source of reactive oxygen species (ROS) generation. Overall, growing evidence now shows that mitochondrial activities and pathways have changed to adapt with different types of leukemia, thus mitochondrial metabolism could be targeted for blood malignancy therapy. This review focuses on the function of mitochondria in LSC of the different leukemia types. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
37. The molecular mechanisms underlying BCR/ABL degradation in chronic myeloid leukemia cells promoted by Beclin1-mediated autophagy
- Author
-
Huang X, Li Y, Shou L, Li L, Chen Z, Ye X, and Qian W
- Subjects
CML ,autophagy ,BCR/ABL ,Beclin1 ,leukemia stem cell ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Xianbo Huang,1,* Ying Li,1,* Lihong Shou,2 Li Li,1 Zhenzhen Chen,3 Xiujin Ye,1 Wenbin Qian1,41Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People’s Republic of China; 2Department of Hematology, The Central Hospital of Huzhou City, Huzhou 313000, People’s Republic of China; 3Department of Hematology, Hangzhou First People’s Hospital, Hangzhou 310003, People’s Republic of China; 4Malignant Lymphoma Diagnosis and Therapy Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, People’s Republic of China*These authors contributed equally to this work Background: The development of drug resistance and the persistence of leukemia stem cells are major obstacles for the use of tyrosine kinase inhibitors (TKIs) in the treatment of chronic myeloid leukemia (CML). The induction of autophagic death in tumor cells represents a new route for leukemia treatment. Our previous study showed that infection of CML cells with oncolytic viruses carrying the autophagy gene Beclin1 downregulated BCR/ABL protein expression and significantly increased the killing effect of the oncolytic viruses on CML cells via autophagy activation. However, the specific molecular mechanisms underlying the regulation of BCR/ABL and Beclin1-dependent CML cell killing remain unclear.Methods: A physical interaction between BCR/ABL and Beclin1 was characterized via GST-pulldown, co-IP and dual-luciferase reporter assays. Cell proliferation was examined via CCK-8 and clone formation assays. The expression levels of the related proteins were measured via Western blotting. Autophagosomes were observed under transmission electron microscopy. Lentiviral vectors carrying Atg7/UVRAG shRNA or the Beclin1 gene were used to modulate the expression levels of the indicated genes. Immunofluorescence were performed to examine colocalization of BCR/ABL and p62/SQSTM1. CD34+,CD38−, cells were isolated from bone marrow samples from CML patients via fluorescence-activated cell sorting.Results: In this study, we observed that Beclin1 directly interacts with BCR/ABL. Beclin1 inhibited the activity of the BCR/ABL promoter to downregulate the level of BCR/ABL protein and to promote the downstream colocalization of p62/SQSTM1 and BCR/ABL to autolysosomes for degradation via activation of the autophagy signaling pathway. In CML cell lines, primary cells and CD34+,CD38−, leukemia stem cells, Beclin1 overexpression significantly inhibited cell growth and proliferation and induced autophagy.Conclusion: Taken together, our results suggest that autophagy induction via Beclin1 overexpression might offer new approaches for treating TKI-resistant CML and for promoting the clearance of leukemia stem cells, both of which have important clinical implications.Keywords: CML, autophagy, BCR/ABL, Beclin1, leukemia stem cell
- Published
- 2019
38. Wnt Signaling Pathway and Chronic Myelogenous Leukemia
- Author
-
HOU Yan, LI Wenqian, FENG Jianming, AI Guo, XIE Youbang, HUA Qingcuo, and ZHAO Changming
- Subjects
chronic myelogenous leukemia ,wnt ,leukemia stem cell ,tki ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
As a type of cancer that starts in certain blood-forming cells of the bone marrow, chronic myeloid leukemia (CML) is caused by the formation of an abnormal gene called BCR-ABL1. The targeted medicine TKI for BCR-ABL1 has great effect on treating CML: however, TKI is not able to completely remove the leukemia stem cells (LSCs) in CML patients, which will lead to drug resistance and leukemia relapse. With the application of Wnt signaling pathway in regulating stem cell function, many studies have found that it also plays an important role in treating CML. In this paper, we reviewed the impact of Wnt signaling pathway on CML therapy.
- Published
- 2019
- Full Text
- View/download PDF
39. Preclinical Evaluation of the Multiple Tyrosine Kinases Inhibitor Anlotinib in Leukemia Stem Cells
- Author
-
Yuelong Jiang, Long Liu, Yirong Jiang, Zhifeng Li, Liying Feng, Xinguo Zhuang, Zhijuan Lin, Qiuling Chen, Guoshu Chen, Jixiang He, Guowei Li, Jie Zha, and Bing Xu
- Subjects
acute myeloid leukemia ,leukemia stem cell ,anlotinib ,JAK ,STAT3 ,chemoresistance ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Leukemia stem cells (LSCs) constitute the critical barrier to the cure of acute myeloid leukemia (AML) due to their chemoresistance and immune evasion property. Herein, the role of anlotinib, a multiple tyrosine kinase inhibitor, in killing LSCs and regulating chemoresistance and immune evasion was explored. Anlotinib treatment induced apoptosis of LSC-like cells as well as primary AML LSCs, while sparing the normal mononuclear cells in vitro. Moreover, anlotinib could impair the regeneration capacity of LSCs in the patient-derived leukemia xenograft mouse model. Mechanistically, anlotinib inhibited phosphorylation of c-kit, JAK2/STAT3, and STAT5, and downregulated STAT3 and STAT5 expression. In addition, anlotinib downregulated the anti-apoptotic proteins Bcl-2 and Bcl-xL, and upregulated Bax, thereby enhancing the sensitivity of LSCs to idarubicin in vitro. Intriguingly, anlotinib could also partially rescue the interferon-g production of T cells cocultured with LSCs by downregulating PD-L1 expression. In conclusion, anlotinib showed anti-LSC activity and the potential to enhance the sensitivity to idarubicin and inhibit the immunosuppressive feature of LSCs via JAK2/STAT signaling pathway downregulation in the preclinical study. Our results provided a rational basis for combinatory strategies involving anlotinib and chemotherapy or immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
40. Leukemia stem cell-bone marrow microenvironment interplay in acute myeloid leukemia development.
- Author
-
Yao, Yiyi, Li, Fenglin, Huang, Jiansong, Jin, Jie, and Wang, Huafeng
- Subjects
- *
ACUTE myeloid leukemia , *LEUKEMIA , *BONE marrow , *CANCER chemotherapy , *OVERALL survival - Abstract
Despite the advances in intensive chemotherapy regimens and targeted therapies, overall survival (OS) of acute myeloid leukemia (AML) remains unfavorable due to inevitable chemotherapy resistance and high relapse rate, which mainly caused by the persistence existence of leukemia stem cells (LSCs). Bone marrow microenvironment (BMM), the home of hematopoiesis, has been considered to play a crucial role in both hematopoiesis and leukemogenesis. When interrupted by the AML cells, a malignant BMM formed and thus provided a refuge for LSCs and protecting them from the cytotoxic effects of chemotherapy. In this review, we summarized the alterations in the bidirectional interplay between hematopoietic cells and BMM in the normal/AML hematopoietic environment, and pointed out the key role of these alterations in pathogenesis and chemotherapy resistance of AML. Finally, we focused on the current potential BMM-targeted strategies together with future prospects and challenges. Accordingly, while further research is necessary to elucidate the underlying mechanisms behind LSC–BMM interaction, targeting the interaction is perceived as a potential therapeutic strategy to eradicate LSCs and ultimately improve the outcome of AML. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
41. p27kip1 maintains a subset of leukemia stem cells in the quiescent state in murine MLL‐leukemia
- Author
-
Zhang, Jun, Seet, Christopher S, Sun, Clare, Li, Jing, You, Dewen, Volk, Andrew, Breslin, Peter, Li, Xingyu, Wei, Wei, Qian, Zhijian, Zeleznik-Le, Nancy J, Zhang, Zhou, and Zhang, Jiwang
- Subjects
Stem Cell Research - Nonembryonic - Human ,Pediatric Cancer ,Hematology ,Stem Cell Research ,Genetics ,Rare Diseases ,Childhood Leukemia ,Cancer ,Pediatric ,Stem Cell Research - Nonembryonic - Non-Human ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Biomarkers ,Tumor ,CD11b Antigen ,Cell Cycle Checkpoints ,Cyclin-Dependent Kinase Inhibitor p27 ,Drug Resistance ,Neoplasm ,Gene Expression Regulation ,Leukemic ,Homeodomain Proteins ,Leukemia ,Biphenotypic ,Acute ,Leukemia ,Myeloid ,Acute ,Mice ,Mice ,Knockout ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Neoplasm Proteins ,Neoplastic Stem Cells ,Proto-Oncogene Proteins c-kit ,Proto-Oncogene Proteins c-myc ,MLL-leukemia ,p27(kip1) ,Leukemia stem cell ,Quiescence ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
MLL (mixed-lineage leukemia)-fusion genes induce the development of leukemia through deregulation of normal MLL target genes, such as HOXA9 and MEIS1. Both HOXA9 and MEIS1 are required for MLL-fusion gene-induced leukemogenesis. Co-expression of HOXA9 and MEIS1 induces acute myeloid leukemia (AML) similar to that seen in mice in which MLL-fusion genes are over-expressed. p27(kip1) (p27 hereafter), a negative regulator of the cell cycle, has also been defined as an MLL target, the expression of which is up-regulated in MLL leukemic cells (LCs). To investigate whether p27 plays a role in the pathogenesis of MLL-leukemia, we examined the effects of p27 deletion (p27(-/-)) on MLL-AF9 (MA9)-induced murine AML development. HOXA9/MEIS1 (H/M)-induced, p27 wild-type (p27(+/+)) and p27(-/-) AML were studied in parallel as controls. We found that LCs from both MA9-AML and H/M-AML can be separated into three fractions, a CD117(-)CD11b(hi) differentiated fraction as well as CD117(+)CD11b(hi) and CD117(+)CD11b(lo), two less differentiated fractions. The CD117(+)CD11b(lo) fraction, comprising only 1-3% of total LCs, expresses higher levels of early hematopoietic progenitor markers but lower levels of mature myeloid cell markers compared to other populations of LCs. p27 is expressed and is required for maintaining the quiescent and drug-resistant states of the CD117(+)CD11b(lo) fraction of MA9-LCs but not of H/M-LCs. p27 deletion significantly compromises the leukemogenic capacity of CD117(+)CD11b(lo) MA9-LCs by reducing the frequency of leukemic stem cells (LSCs) but does not do so in H/M-LCs. In addition, we found that p27 is highly expressed and required for cell cycle arrest in the CD117(-)CD11b(hi) fraction in both types of LCs. Furthermore, we found that c-Myc expression is required for maintaining LCs in an undifferentiated state independently of proliferation. We concluded that p27 represses the proliferation of LCs, which is specifically required for maintaining the quiescent and drug-resistant states of a small subset of MA9-LSCs in collaboration with the differentiation blockage function of c-Myc.
- Published
- 2013
42. [Clinical characteristics and prognosis of childhood acute lymphoblastic leukemia with CD123 expression].
- Author
-
Zhu KF, Li HJ, Qiao CF, Wang LF, Wu PJ, Chen Y, and Tian X
- Subjects
- Humans, Male, Female, Child, Child, Preschool, Prognosis, Retrospective Studies, Infant, Adolescent, Interleukin-3 Receptor alpha Subunit analysis, Interleukin-3 Receptor alpha Subunit genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality
- Abstract
Objectives: To investigate the expression of CD123 in children with acute lymphoblastic leukemia (ALL) and its effect on the clinical characteristics and prognosis of children with B-lineage acute lymphoblastic leukemia (B-ALL)., Methods: A retrospective analysis was conducted on the clinical data of 251 children with ALL who were admitted to the Department of Hematology and Oncology, Children's Hospital of Kunming Medical University, from December 2019 to June 2022. According to the expression of CD123 at initial diagnosis, the children were divided into CD123
+ group and CD123- group, and the two groups were compared in terms of clinical characteristics and treatment outcome. The factors influencing the prognosis were analyzed., Results: Among the 251 children with ALL, there were 146 children (58.2%) in the CD123+ group. The B-ALL group had a significantly higher positive expression rate of CD123 than the acute T lymphocyte leukemia group ( P <0.05). Compared with the CD123- group, the CD123+ group had significantly lower peripheral blood leukocyte count and percentage of juvenile cells and a significantly higher proportion of children with high hyperdiploid karyotype or an age of 1-10 years, with a relatively low proportion of children with E2A-PBX1 fusion gene ( P <0.05). The multivariate Cox proportional-hazards regression model analysis showed that compared with the >10 years group, the 1-10 years group had a significantly higher overall survival rate ( P <0.05), and compared with the high risk group, the moderate risk group had a significantly higher event-free survival rate in children with B-ALL ( P <0.05)., Conclusions: CD123 is widely expressed in children with B-ALL, and positive expression of CD123 might be an indicator for good prognosis in children with B-ALL, which is of great significance for evaluating the efficacy of remission induction therapy and survival prognosis of children with B-ALL.- Published
- 2024
- Full Text
- View/download PDF
43. Leukemia Stem Cell Release From the Stem Cell Niche to Treat Acute Myeloid Leukemia
- Author
-
Alicia Villatoro, Joanna Konieczny, Vincent Cuminetti, and Lorena Arranz
- Subjects
leukemia stem cell ,hematopoietic stem cell ,homing ,mobilization ,adhesion ,hematopoietic stem cell niche ,Biology (General) ,QH301-705.5 - Abstract
Acute myeloid leukemia (AML) is a heterogeneous, complex, and deadly disease, whose treatment has hardly evolved for decades and grounds on the use of intensive chemotherapy regimens. Chemotherapy helps reduce AML bulk, but promotes relapse in the long-run by selection of chemoresistant leukemia stem cells (LSC). These may diversify and result in progression to more aggressive forms of AML. In vivo models suggest that the bone marrow stem cell niche helps LSC stay dormant and protected from chemotherapy. Here, we summarize relevant changes in stem cell niche homing and adhesion of AML LSC vs. healthy hematopoietic stem cells, and provide an overview of clinical trials aiming at targeting these processes for AML treatment and future directions within this field. Promising results with various non-mutation-targeted novel therapies directed to LSC eradication via interference with their anchoring to the stem cell niche have encouraged on-going or future advanced phase III clinical trials. In the coming years, we may see a shift in the focus of AML treatment to LSC-directed therapies if the prospect of improved cure rates holds true. In the future, AML treatment should lean toward personalized therapies using combinations of these compounds plus mutation-targeted agents and/or targeted delivery of chemotherapy, aiming at LSC eradication with reduced side effects.
- Published
- 2020
- Full Text
- View/download PDF
44. All-trans retinoic acid in non-promyelocytic acute myeloid leukemia: driver lesion dependent effects on leukemic stem cells.
- Author
-
Nguyen, Chi H., Grandits, Alexander M., Purton, Louise E., Sill, Heinz, and Wieser, Rotraud
- Subjects
PRELEUKEMIA ,ACUTE myeloid leukemia ,ACUTE promyelocytic leukemia ,STEM cells ,TRETINOIN ,HEMATOLOGIC malignancies - Abstract
Acute myeloid leukemia (AML) is an aggressive, often fatal hematopoietic malignancy. All-trans retinoic acid (atRA), one of the first molecularly targeted drugs in oncology, has greatly improved the outcome of a subtype of AML, acute promyelocytic leukemia (APL). In contrast, atRA has so far provided little therapeutic benefit in the much larger group of patients with non-APL AML. Attempts to identify genetically or molecularly defined subgroups of patients that may respond to atRA have not yielded consistent results. Since AML is a stem cell-driven disease, understanding the effectiveness of atRA may require an appreciation of its impact on AML stem cells. Recent studies reported that atRA decreased stemness of AML with an FLT3-ITD mutation, yet increased it in AML1-ETO driven or EVI1-overexpressing AML. This review summarizes the role of atRA in normal hematopoiesis and in AML, focusing on its impact on AML stem cells. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
45. WT1 facilitates the self-renewal of leukemia-initiating cells through the upregulation of BCL2L2: WT1-BCL2L2 axis as a new acute myeloid leukemia therapy target.
- Author
-
Zhou, Bin, Jin, Xianghong, Jin, Weiwei, Huang, Xingzhou, Wu, Yanfei, Li, Haiying, Zhu, Weijian, Qin, Xiaoyi, Ye, Haige, and Gao, Shenmeng
- Subjects
- *
ACUTE myeloid leukemia , *MOUSE leukemia , *PROTEASOMES , *STEM cells , *RNA sequencing , *CELLS , *UBIQUITINATION - Abstract
Background: Overexpression of Wilms' tumor-1 (WT1) transcription factor facilitates proliferation in acute myeloid leukemia (AML). However, whether WT1 is enriched in the leukemia-initiating cells (LICs) and leukemia stem cells (LSCs) and facilitates the self-renewal of LSCs remains poorly understood.Methods: MLL-AF9-induced murine leukemia model was used to evaluate the effect of knockdown of wt1 on the self-renewal ability of LSC. RNA sequencing was performed on WT1-overexpressing cells to select WT1 targets. Apoptosis and colony formation assays were used to assess the anti-leukemic potential of a deubiquitinase inhibitor WP1130. Furthermore, NOD/SCID-IL2Rγ (NSG) AML xenotransplantation and MLL-AF9-induced murine leukemia models were used to evaluate the anti-leukemogenic potential of WP1130 in vivo.Results: We found that wt1 is highly expressed in LICs and LSCs and facilitates the maintenance of leukemia in a murine MLL-AF9-induced model of AML. WT1 enhanced the self-renewal of LSC by increasing the expression of BCL2L2, a member of B cell lymphoma 2 (BCL2) family, by direct binding to its promoter region. Loss of WT1 impaired self-renewal ability in LSC and delayed the progression of leukemia. WP1130 was found to modify the WT1-BCL2L2 axis, and WP1130-induced anti-leukemic activity was mediated by ubiquitin proteasome-mediated destruction of WT1 protein. WP1130 induced apoptosis and decreased colony formation abilities of leukemia cells and prolonged the overall survival in the THP1-based xenograft NSG mouse model. WP1130 also decreased the frequency of LSC and prolonged the overall survival in MLL-AF9-induced murine leukemia model. Mechanistically, WP1130 induced the degradation of WT1 by positively affecting the ubiquitination of WT1 protein.Conclusions: Our results indicate that WT1 is required for the development of AML. WP1130 exhibits anti-leukemic activity by inhibiting the WT1-BCL2L2 axis, which may represent a new acute myeloid leukemia therapy target. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
46. Aberrant cytokine signaling in leukemia
- Author
-
Van Etten, RA
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Hematology ,Childhood Leukemia ,Cancer ,Pediatric ,Pediatric Cancer ,Rare Diseases ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Animals ,Cell Transformation ,Neoplastic ,Cytokines ,Humans ,Leukemia ,Signal Transduction ,tyrosine kinase ,kinase inhibitor ,leukemia stem cell ,Clinical Sciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Abnormalities of cytokine and growth factor signaling pathways are characteristic of all forms of leukemia: lymphoid and myeloid, acute and chronic. In normal hematopoietic cells, cytokines provide the stimulus for proliferation, survival, self-renewal, differentiation and functional activation. In leukemic cells, these pathways are usurped to subserve critical parts of the malignant program. In this review, our current knowledge of leukemic cell cytokine signaling will be summarized, and some speculations on the significance and implications of these insights will be advanced. A better understanding of aberrant cytokine signaling in leukemia should provide additional targets for the rational therapy of these diseases.
- Published
- 2007
47. CRISPR Screens Identify Candidate Therapeutic Targets in Leukemia
- Author
-
Rodriguez Zabala, Maria and Rodriguez Zabala, Maria
- Abstract
Acute myeloid leukemia (AML) is a complex hematological malignancy marked by proliferation of immature myeloid cells with a dismal 5-year survival. A major challenge is the persistence of leukemia stem cells (LSCs) after standard treatment, leading to relapse. This thesis employs in vivo CRISPR/Cas9 screening to investigate critical AML and LSC molecular mechanisms, interrogating the dependancies on cell surface receptors for use as potential therapies. In our initial study (Article I), we identify the chemokine receptor CXCR4 as a crucial dependency of AML cell growth and survival. Using a murine model of AML driven by MLL::AF9, we find that CXCR4 loss triggers oxidative stress and differentiation in vivo, with CXCL12 ligand signaling being non-essential for leukemia development.Expanding our study to interrogate nearly one thousand cell surface receptors, we identify three additional AML dependencies. Among these, GLUT1, a primary cellular glucose transporter, emerges as a key regulator of energy metabolism, driving MLL::AF9 LSC survival (Article II). Inhibition of GLUT1 suppresses cellular bioenergetics, prompting autophagy as a metabolic adaptation. Notably, dual inhibition of GLUT1 and oxidative phosphorylation effectively eliminates human AML cells, especially for the RUNX1-mutated AML subtype.Furthermore, our research also reveals iron metabolism as another critical AML dependency (Article III). We found that disrupting iron uptake through genetic knockdown of Tfrc, encoding the transferrin receptor (TFR1), suppresses leukemia development in a p53-dependent manner, leading to transcriptional repression of antioxidant defense and mitochondrial respiration pathways. Patient-derived AML cells were selectively targeted upon iron chelation treatment. Additionally, our work uncovers the role of H2K1 in evading NK cell-mediated immune surveillance in vivo through disruption of NK cell maturation and activation (Article IV). Consistent with this finding, H2k1 disrupt
- Published
- 2023
48. Prospective monitoring of chronic myeloid leukemia patients from the time of TKI discontinuation: the fate of peripheral blood CD26+ leukemia stem cells
- Author
-
Pacelli, P., Santoni, A., Sicuranza, A., Abruzzese, E., Giai, V., Crugnola, M., Annunziata, M., Galimberti, S., Iurlo, A., Luciano, L., Sora', Federica, Fava, C., Bestoso, E., Marzano, C., Cartocci, A., Defina, M., Sammartano, V., Cencini, E., Raspadori, D., Bocchia, M., Sora' F. (ORCID:0000-0002-9607-5298), Pacelli, P., Santoni, A., Sicuranza, A., Abruzzese, E., Giai, V., Crugnola, M., Annunziata, M., Galimberti, S., Iurlo, A., Luciano, L., Sora', Federica, Fava, C., Bestoso, E., Marzano, C., Cartocci, A., Defina, M., Sammartano, V., Cencini, E., Raspadori, D., Bocchia, M., and Sora' F. (ORCID:0000-0002-9607-5298)
- Abstract
Introduction: In chronic myeloid leukemia (CML), about half of the patients achieving a deep and stable molecular response with tyrosine kinase inhibitors (TKIs) may discontinue TKI treatment without disease recurrence. As such, treatment-free remission (TFR) has become an ambitious goal of treatment. Given the evidence that deepness and duration of molecular response are necessary but not sufficient requisites for a successful TFR, additional biological criteria are needed to identify CML patients suitable for efficacious discontinuation. Leukemia stem cells (LSCs) are supposed to be the reservoir of the disease. Previously, we demonstrated that residual circulating CD34+/CD38-/CD26+ LSCs were still detectable in a consistent number of CML patients during TFR. Methods: CML LSCs could be easily identified by flow-cytometry as they express the CD34+/CD38-/CD26+ phenotype. In this study, we explored the role of these cells and their correlation with molecular response in a cohort of 109 consecutive chronic phase CML patients prospectively monitored from the time of TKI discontinuation. Results: After a median observation time of 33 months from TKI discontinuation, 38/109 (35%) patients failed TFR after a median time of 4 months, while 71/109 (65%) patients are still in TFR. At TKI discontinuation, peripheral blood CD26+LSCs were undetectable in 48/109 (44%) patients and detectable in 61/109 (56%). No statistically significant correlation between detectable/undetectable CD26+LSCs and the rate of TFR loss was found (p = 0.616). The incidence of TFR loss based on the type of TKI treatment was statistically significant for imatinib treatment compared to that of nilotinib (p = 0.039). Exploring the behavior of CD26+LSCs during TFR, we observed fluctuating values that were very variable between patients, and they were not predictive of TFR loss. Discussion: Up to date, our results confirm that CD26+LSCs are detectable at the time of TKI discontinuation and during
- Published
- 2023
49. Leukemia stem cells promote chemoresistance by inducing downregulation of lumican in mesenchymal stem cells.
- Author
-
Yu, Zhen, Liu, Lin, Shu, Qiang, Li, Dong, and Wang, Ran
- Subjects
- *
MESENCHYMAL stem cells , *STEM cells , *LEUKEMIA , *LYMPHOBLASTIC leukemia , *WESTERN immunoblotting - Abstract
Leukemia stem cells (LSCs) are responsible for therapeutic failure and relapse of acute lymphoblastic leukemia. As a result of the interplay between LSCs and bone marrow mesenchymal stem cells (BM-MSCs), cancer cells may escape from chemotherapy and immune surveillance, thereby promoting leukemia progress and relapse. The present study identified that the crosstalk between LSCs and BM-MSCs may contribute to changes of immune phenotypes and expression of hematopoietic factors in BM-MSCs. Furthermore, Illumina Genome Analyzer/Hiseq 2000 identified 7 differentially expressed genes between BM-MSCsLSC and BM-MSCs. The Illumina sequencing results were further validated by reverse transcription-quantitative polymerase chain reaction. Following LSC simulation, 2 genes were significantly upregulated, whereas the remaining 2 genes were significantly downregulated in MSCs. The most remarkable changes were identified in the expression levels of lumican (LUM) gene. These results were confirmed by western blot analysis. In addition, decreased LUM expression led to decreased apoptosis, and promoted chemoresistance to VP-16 in Nalm-6 cells. These results suggest that downregulation of LUM expression in BM-MSCs contribute to the anti-apoptotic properties and resistance to chemotherapy in LSCs. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
50. Six1 regulates leukemia stem cell maintenance in acute myeloid leukemia.
- Author
-
Chu, Yajing, Chen, Yangpeng, Li, Mengke, Shi, Deyang, Wang, Bichen, Lian, Yu, Cheng, Xuelian, Wang, Xiaomin, Xu, Mingjiang, Cheng, Tao, Shi, Jun, and Yuan, Weiping
- Abstract
Molecular genetic changes in acute myeloid leukemia (AML) play crucial roles in leukemogenesis, including recurrent chromosome translocations, epigenetic/spliceosome mutations and transcription factor aberrations. Six1, a transcription factor of the Sine oculis homeobox (Six) family, has been shown to transform normal hematopoietic progenitors into leukemia in cooperation with Eya. However, the specific role and the underlying mechanism of Six1 in leukemia maintenance remain unexplored. Here, we showed increased expression of SIX1 in AML patients and murine leukemia stem cells (c‐Kit+ cells, LSCs). Importantly, we also observed that a higher level of Six1 in human patients predicts a worse prognosis. Notably, knockdown of Six1 significantly prolonged the survival of MLL‐AF9‐induced AML mice with reduced peripheral infiltration and tumor burden. AML cells from Six1‐knockdown (KD) mice displayed a significantly decreased number and function of LSC, as assessed by the immunophenotype, colony‐forming ability and limiting dilution assay. Further analysis revealed the augmented apoptosis of LSC and decreased expression of glycolytic genes in Six1 KD mice. Overall, our data showed that Six1 is essential for the progression of MLL‐AF9‐induced AML via maintaining the pool of LSC. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.