1
|
Lu J, Lv L, Tian X, Li Z, Ma Y, Li N, Wang J, Wang G, Zeng Y, Zhang W, Xu J, Liang A. CD37 regulates the self-renewal of leukemic stem cells via integrin-mediated signaling in acute myeloid leukemia. Stem Cell Reports 2025; 20:102476. [PMID: 40250439 DOI: 10.1016/j.stemcr.2025.102476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/20/2025] Open
Abstract
Leukemic stem cells (LSCs) are a small subset of leukemia cells that drive leukemia initiation and maintenance. Herein, we report that CD37, a member of transmembrane 4 superfamily (TM4SF), regulates the survival of acute myeloid leukemia (AML) cells as well as the self-renewal of AML LSCs. The downregulation of CD37 retarded proliferation and increased apoptosis in human AML cell lines THP-1 and OCI-AML2. Deficiency of CD37 in vivo had a minimal effect on normal hematopoiesis but significantly impeded leukemia maintenance and propagation, which led to increased apoptosis and decreased cell cycle entry in AML blasts as well as impaired colony formation and declined frequency of AML LSCs in the serial transplantation. Furthermore, CD37 interacted with integrin α4β7 and activated the phosphatidylinositol 3-kinase (PI3K)-AKT pathway mediated by integrin signaling. Our study provides novel insights for targeted therapy of AML, indicating CD37 as a safe and effective target for immunotherapy.
Collapse
Affiliation(s)
- Jinyuan Lu
- Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, P.R. China; Department of Hematology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200333, P.R. China
| | - Lixin Lv
- Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, P.R. China; Department of Hematology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200333, P.R. China
| | - Xiaoxue Tian
- Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, P.R. China; Department of Hematology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200333, P.R. China
| | - Zheng Li
- Department of Hematology, Renji Hospital, Shanghai Jiaotong University School of Medicine, 160 Pujian Road, Shanghai 201112, P.R. China
| | - Yuting Ma
- Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, P.R. China; Department of Hematology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200333, P.R. China
| | - Nannan Li
- Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, P.R. China; Department of Hematology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200333, P.R. China
| | - Jian Wang
- Tongji University School of Medicine, 500 Zhennan Road, Shanghai 200331, P.R. China; Stem Cell Research Center, East Hospital, Tongji University School of Medicine, Shanghai 200331, P.R. China
| | - Guangming Wang
- Stem Cell Research Center, East Hospital, Tongji University School of Medicine, Shanghai 200331, P.R. China
| | - Yu Zeng
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200333, P.R. China
| | - Wenjun Zhang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200333, P.R. China
| | - Jun Xu
- Stem Cell Research Center, East Hospital, Tongji University School of Medicine, Shanghai 200331, P.R. China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai 200333, P.R. China.
| |
Collapse
|
2
|
Nguyen NTK, Yao H, Hosokawa K, Esaki Y, Yuta R, Adachi S, Arai F. Acute myeloid leukemia stem cells remodel the bone marrow niche via TGF-β-activated Alcam + bone lining cells, creating a self-sustaining environment. Leukemia 2025:10.1038/s41375-025-02640-4. [PMID: 40360881 DOI: 10.1038/s41375-025-02640-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Affiliation(s)
- Ngan Thi Kim Nguyen
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hisayuki Yao
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Hosokawa
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki Esaki
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryosuke Yuta
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shunichi Adachi
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumio Arai
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
3
|
Cao S, Wang Q, Zhu G. From Chemotherapy to Targeted Therapy: Unraveling Resistance in Acute Myeloid Leukemia Through Genetic and Non-Genetic Insights. Int J Mol Sci 2025; 26:4005. [PMID: 40362245 PMCID: PMC12071668 DOI: 10.3390/ijms26094005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Acute myeloid leukemia (AML) is a devastating disease characterized by extensive inter-patient and intra-patient heterogeneity. Despite the introduction of intensive chemotherapy in the 1970s as the standard treatment, the development of mechanism-based targeted therapies since 2017 has been broadening the therapeutic landscape. However, both chemotherapy and targeted therapies continue to face the challenges of primary and secondary resistance. This review summarizes the mechanisms underlying resistance to chemotherapy and targeted therapies in AML and discusses the opportunities and challenges brought by the transition from chemotherapy to precision medicine.
Collapse
Affiliation(s)
| | | | - Ganqian Zhu
- School of Biomedical Sciences, Hunan University, Changsha 410082, China; (S.C.); (Q.W.)
| |
Collapse
|
4
|
Dong J, Konopleva M. Preclinical targeting of leukemia-initiating cells in the development future biologics for acute myeloid leukemia. Expert Opin Ther Targets 2025; 29:223-237. [PMID: 40304258 DOI: 10.1080/14728222.2025.2500417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Leukemia-initiating cells (LICs) are a critical subset of cells driving acute myeloid leukemia (AML) relapse and resistance to therapy. They possess unique properties, including metabolic, epigenetic, and microenvironmental dependencies, making them promising therapeutic targets. AREAS COVERED This review summarizes preclinical advances in targeting AML LICs, including strategies to exploit metabolic vulnerabilities, such as the reliance on oxidative phosphorylation (OXPHOS), through the use of mitochondrial inhibitors; target epigenetic regulators like DOT1L (Disruptor of Telomeric Silencing 1-like) to disrupt LIC survival mechanisms; develop immunotherapies, including CAR (chimeric antigen receptor) T-cell therapy, and bispecific antibodies; and disrupt LIC interactions with the bone marrow microenvironment by inhibiting supportive niches. EXPERT OPINION LIC-targeted therapies hold significant promise for revolutionizing AML treatment by reducing relapse rates and improving long-term outcomes. However, challenges such as LIC heterogeneity, therapy resistance, and associated toxicity persist. Recent studies have illuminated the distinct biological characteristics of LICs, advancing our understanding of their behavior and vulnerabilities. These insights offer new opportunities to target LICs at earlier disease stages and to explore combination therapies with other targeted treatments, ultimately enhancing therapeutic efficacy and improving patient outcomes.
Collapse
Affiliation(s)
- Jiaxin Dong
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marina Konopleva
- Department of Medicine (Oncology), Blood Cancer Institute, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
5
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
6
|
Sakoda T, Kikushige Y, Irifune H, Kawano G, Harada T, Semba Y, Hayashi M, Shima T, Mori Y, Eto T, Kamimura T, Iwasaki H, Ogawa R, Yoshimoto G, Kato K, Maeda T, Miyamoto T, Akashi K. TIM-3 marks measurable residual leukemic stem cells responsible for relapse after allogeneic stem cell transplantation. Cancer Sci 2025; 116:698-709. [PMID: 39726280 PMCID: PMC11875787 DOI: 10.1111/cas.16431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
In this study, we investigated the measurable residual leukemic stem cell (MR-LSC) population after allogeneic stem cell transplantation (allo-SCT) for high-risk acute myeloid leukemia (AML), utilizing T-cell immunoglobulin mucin-3 (TIM-3) expression as a functional marker of AML leukemic stem cells (LSCs). Analysis of the CD34+CD38- fraction of bone marrow cells immediately after achievement of engraftment revealed the presence of both TIM-3+LSCs and TIM-3- donor hematopoietic stem cells (HSCs) at varying ratios. Genetic analysis confirmed that TIM-3+ cells harbored patient-specific mutations identical to those found in AML clones, whereas TIM-3- cells did not, indicating that TIM-3+CD34+CD38- cells represent residual AML LSCs. In 92 allo-SCT occasions involving 83 AML patients, we enumerated the frequencies of TIM-3+LSCs immediately after achieving hematologic complete remission with complete donor cell chimerism. Notably, only 22.2% of patients who achieved a TIM-3+MR-LSClow status (<60%) experienced relapse, with a median event-free survival (EFS) of 1581 days (median follow-up duration was 2177 days among event-free survivors). Conversely, 87.5% of patients with TIM-3+MR-LSCint/high (≥60%) relapsed, with a median EFS of 140.5 days. Furthermore, MR-LSC status emerged as a significant independent risk factor for relapse (hazard ratio, 8.56; p < 0.0001), surpassing the impact of patient disease status prior to allo-SCT, including failure to achieve complete remission (hazard ratio, 1.98; p = 0.048). These findings suggest that evaluating TIM-3+ MR-LSCs immediately after engraftment, which reflects the competitive reconstitution of residual TIM-3+ LSCs and donor HSCs, may be valuable for predicting outcomes in AML patients undergoing allo-SCT.
Collapse
MESH Headings
- Humans
- Hepatitis A Virus Cellular Receptor 2/metabolism
- Hepatitis A Virus Cellular Receptor 2/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Male
- Middle Aged
- Female
- Adult
- Hematopoietic Stem Cell Transplantation
- Transplantation, Homologous
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplasm, Residual
- Young Adult
- Aged
- Adolescent
- Recurrence
- Disease-Free Survival
- Mutation
Collapse
Affiliation(s)
- Teppei Sakoda
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
- Center for Cellular and Molecular MedicineKyushu University HospitalFukuokaJapan
| | - Yoshikane Kikushige
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
- Center for Cellular and Molecular MedicineKyushu University HospitalFukuokaJapan
| | - Hidetoshi Irifune
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
| | - Gentaro Kawano
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
| | - Takuya Harada
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
| | - Yuichiro Semba
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
| | | | - Takahiro Shima
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
| | - Yasuo Mori
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
| | - Tetsuya Eto
- Department of HematologyHamanomachi HospitalFukuokaJapan
| | | | - Hiromi Iwasaki
- Department of HematologyNational Hospital Organisation Kyushu Medical CenterFukuokaJapan
| | - Ryosuke Ogawa
- Department of Hematology/OncologyJapan Community Health Care Organisation Kyushu HospitalKitakyushuJapan
| | - Goichi Yoshimoto
- Department of HematologySaga Prefecture Medical Center KoseikanSagaJapan
| | - Koji Kato
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
| | - Takahiro Maeda
- Division of Precision MedicineKyushu University Faculty of Medicine Graduate School of Medical ScienceFukuokaJapan
| | - Toshihiro Miyamoto
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
| | - Koichi Akashi
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of MedicineFukuokaJapan
- Center for Cellular and Molecular MedicineKyushu University HospitalFukuokaJapan
| |
Collapse
|
7
|
Soleimani Samarkhazan H, Zehtabcheh S, Seraji HR, Beqaj SH, Tayefeh S, Mohammadi MH, Aghaei M. Unveiling the potential of CLL-1: a promising target for AML therapy. Biomark Res 2025; 13:28. [PMID: 39940055 PMCID: PMC11823018 DOI: 10.1186/s40364-025-00738-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
Acute myeloid leukemia (AML) remains a formidable blood cancer, despite recent advances in treatment. A significant challenge persists in improving patient outcomes, particularly in addressing relapse and treatment resistance. Identifying new therapeutic targets is critical for advancing AML therapy. C-type lectin-like molecule-1 (CLL-1) has emerged as a promising therapeutic target in AML. This cell surface receptor is highly expressed on AML blasts and demonstrates stable expression throughout disease progression. CLL-1's consistent presence makes it an ideal candidate for monitoring minimal residual disease (MRD), which is a critical indicator for predicting relapse. Beyond its utility as a diagnostic marker, CLL-1 offers exciting potential in the development of immunotherapies. Emerging strategies, such as CAR-T-cell therapy and antibody-drug conjugates (ADCs), are being investigated to leverage the immune system against CLL-1-expressing AML cells. This review examines the structure, function, and expression patterns of CLL-1 in AML and other hematologic malignancies, providing insights into its role in disease pathogenesis and treatment potential. Exploring CLL-1 as a target for diagnosis, MRD monitoring, and immunotherapy opens new avenues for AML treatment. A deeper understanding of its relationship with AML pathogenesis will aid in the development of targeted therapies, offering hope for improved patient outcomes in the future.
Collapse
Affiliation(s)
- Hamed Soleimani Samarkhazan
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Zehtabcheh
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamideh Rahmani Seraji
- Department of Hematology and Oncology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shamim Tayefeh
- UCLA Immunogenetics Center, Immunogenetics (UIC), 1000 Veteran Ave, Los Angeles, CA, 90024, USA
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mojtaba Aghaei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
8
|
Ahmed F, Zhong J. Advances in DNA/RNA Sequencing and Their Applications in Acute Myeloid Leukemia (AML). Int J Mol Sci 2024; 26:71. [PMID: 39795930 PMCID: PMC11720148 DOI: 10.3390/ijms26010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/24/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive malignancy that poses significant challenges due to high rates of relapse and resistance to treatment, particularly in older populations. While therapeutic advances have been made, survival outcomes remain suboptimal. The evolution of DNA and RNA sequencing technologies, including whole-genome sequencing (WGS), whole-exome sequencing (WES), and RNA sequencing (RNA-Seq), has significantly enhanced our understanding of AML at the molecular level. These technologies have led to the discovery of driver mutations and transcriptomic alterations critical for improving diagnosis, prognosis, and personalized therapy development. Furthermore, single-cell RNA sequencing (scRNA-Seq) has uncovered rare subpopulations of leukemia stem cells (LSCs) contributing to disease progression and relapse. However, widespread clinical integration of these tools remains limited by costs, data complexity, and ethical challenges. This review explores recent advancements in DNA/RNA sequencing in AML and highlights both the potential and limitations of these techniques in clinical practice.
Collapse
Affiliation(s)
| | - Jiang Zhong
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA;
| |
Collapse
|
9
|
Reuvekamp T, Bachas C, Cloos J. Immunophenotypic features of early haematopoietic and leukaemia stem cells. Int J Lab Hematol 2024; 46:795-808. [PMID: 39045906 DOI: 10.1111/ijlh.14348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
Many tumours are organised in a hierarchical structure with at its apex a cell that can maintain, establish, and repopulate the tumour-the cancer stem cell. The haematopoietic stem cell (HSC) is the founder cell for all functional blood cells. Like HSCs, the leukaemia stem cells (LSC) are hypothesised to be the leukaemia-initiating cells, which have features of stemness such as self-renewal, quiescence, and resistance to cytotoxic drugs. Immunophenotypically, CD34+CD38- defines HSCs by adding lineage negativity and CD90+CD45RA-. At which stage of maturation the further differentiation is blocked, determines the type of leukaemia, and determines the immunophenotype of the LSC specific to the leukaemia type. No apparent LSC phenotype has been described in lymphoid leukaemia, and it is debated if a specific acute lymphocytic leukaemia-initiating cell is present, as all cells are capable of engraftment in a secondary mouse model. In chronic lymphocytic leukaemia, a B-cell clone is responsible for uncontrolled proliferation, not a specific LSC. In chronic and acute myeloid leukaemia, LSC is described as CD34+CD38- with the expression of a marker that is aberrantly expressed (LSC marker), such as CD45RA, CD123 or in the case of chronic myeloid leukaemia CD26. In acute myeloid leukaemia, the LSC load had prognostic relevance and might be a biomarker that can be used for monitoring and as an addition to measurable residual disease. However, challenges such as the CD34-negative immunophenotype need to be explored.
Collapse
Affiliation(s)
- Tom Reuvekamp
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Costa Bachas
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Li H, Cao Z, Liu Y, Xue Z, Li Y, Xing H, Xu Y, Gu R, Qiu S, Wei H, Wang M, Rao Q, Wang J. Slow-replicating leukemia cells represent a leukemia stem cell population with high cell-surface CD74 expression. Mol Oncol 2024; 18:2554-2568. [PMID: 38922758 PMCID: PMC11459046 DOI: 10.1002/1878-0261.13690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/25/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Persistence of quiescent leukemia stem cells (LSCs) after treatment most likely contributes to chemotherapy resistance and poor prognosis of leukemia patients. Identification of this quiescent cell population would facilitate eradicating LSCs. Here, using a cell-tracing PKH26 (PKH) dye that can be equally distributed to daughter cells following cell division in vivo, we identify a label-retaining slow-cycling leukemia cell population from AML1-ETO9a (AE9a) leukemic mice. We find that, compared with cells not maintaining PKH-staining, a higher proportion of PKH-retaining cells are in G0 phase, and PKH-retaining cells exhibit increased colony formation ability and leukemia initiation potential. In addition, PKH-retaining cells possess high chemo-resistance and are more likely to be localized to the endosteal bone marrow region. Based on the transcriptional signature, HLA class II histocompatibility antigen gamma chain (Cd74) is highly expressed in PKH-retaining leukemia cells. Furthermore, cell surface CD74 was identified to be highly expressed in LSCs of AE9a mice and CD34+ human leukemia cells. Compared to Lin-CD74- leukemia cells, Lin-CD74+ leukemia cells of AE9a mice exhibit higher stemness properties. Collectively, our findings reveal that the identified slow-cycling leukemia cell population represents an LSC population, and CD74+ leukemia cells possess stemness properties, suggesting that CD74 is a candidate LSC surface marker.
Collapse
Affiliation(s)
- Huan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Zhijie Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Yiming Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Zhenya Xue
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Yishuang Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Runxia Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Shaowei Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Hui Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| |
Collapse
|
11
|
Boutzen H, Murison A, Oriecuia A, Bansal S, Arlidge C, Wang JCY, Lupien M, Kaufmann KB, Dick JE. Identification of leukemia stem cell subsets with distinct transcriptional, epigenetic and functional properties. Leukemia 2024; 38:2090-2101. [PMID: 39169113 PMCID: PMC11436360 DOI: 10.1038/s41375-024-02358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024]
Abstract
The leukemia stem cell (LSC) compartment is a complex reservoir fueling disease progression in acute myeloid leukemia (AML). The existence of heterogeneity within this compartment is well documented but prior studies have focused on genetic heterogeneity without being able to address functional heterogeneity. Understanding this heterogeneity is critical for the informed design of therapies targeting LSC, but has been hampered by LSC scarcity and the lack of reliable cell surface markers for viable LSC isolation. To overcome these challenges, we turned to the patient-derived OCI-AML22 cell model. This model includes functionally, transcriptionally and epigenetically characterized LSC broadly representative of LSC found in primary AML samples. Focusing on the pool of LSC, we used an integrated approach combining xenograft assays with single-cell analysis to identify two LSC subtypes with distinct transcriptional, epigenetic and functional properties. These LSC subtypes differed in depth of quiescence, differentiation potential, repopulation capacity, sensitivity to chemotherapy and could be isolated based on CD112 expression. A majority of AML patient samples had transcriptional signatures reflective of either LSC subtype, and some even showed coexistence within an individual sample. This work provides a framework for investigating the LSC compartment and designing combinatorial therapeutic strategies in AML.
Collapse
Affiliation(s)
- Héléna Boutzen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
| | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Alexa Oriecuia
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Suraj Bansal
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Christopher Arlidge
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5S 1A4, Canada
| | - Kerstin B Kaufmann
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 0A3, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
12
|
Wang YQ, Ren Y, Gale RP, Niu LT, Huang XJ. Sphingosine-1 phosphate receptor 1 (S1PR1) expression maintains stemness of acute myeloid leukemia stem cells. Cancer Lett 2024; 600:217158. [PMID: 39111385 DOI: 10.1016/j.canlet.2024.217158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/24/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Acute myeloid leukemia (AML) arises from leukemia stem cells (LSCs) and is maintained by cells which have acquired features of stemness. We compared transcription profiles of AML cells with/without stem cell features defined as in vitro clonogenicity and serial engraftment in immune-deficient mice xenograft model. We used multi-parameter flow cytometry (MPFC) to separate CD34+ bone marrow-derived leukemia cells into sphingosine-1 phosphate receptor 1 (S1PR1)+ and S1PR1- fractions. Cells in the S1PR1+ fraction demonstrated significantly higher clonogenicity and higher engraftment potential compared with those in the S1PR1- fraction. In contrast, CD34+ bone marrow cells from normal samples showed reduced clonogenicity in the S1PR1+ fraction compared with the S1PR1- fraction. Inhibition of S1PR1 expression in an AML cell line reduced the colony-forming potential of KG1 cells. Transcriptomic analyses and rescue experiments indicated PI3K/AKT pathway and MYBL2 are downstream mediators of S1PR1-associated stemness. These findings implicate S1PR1 as a functional biomarker of LSCs and suggest its potential as a therapeutic target in AML treatment.
Collapse
Affiliation(s)
- Yu-Qing Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yue Ren
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Robert Peter Gale
- Centre for Hematology Research, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Li-Ting Niu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematological Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
13
|
Grenier JMP, Testut C, Bal M, Bardin F, De Grandis M, Gelsi-Boyer V, Vernerey J, Delahaye M, Granjeaud S, Zemmour C, Spinella JF, Chavakis T, Mancini SJC, Boher JM, Hébert J, Sauvageau G, Vey N, Schwaller J, Hospital MA, Fauriat C, Aurrand-Lions M. Genetic deletion of JAM-C in preleukemic cells rewires leukemic stem cell gene expression program in AML. Blood Adv 2024; 8:4662-4678. [PMID: 38954834 PMCID: PMC11402138 DOI: 10.1182/bloodadvances.2023011747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 05/23/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
ABSTRACT The leukemic stem cell (LSC) score LSC-17 based on a stemness-related gene expression signature is an indicator of poor disease outcome in acute myeloid leukemia (AML). However, it is not known whether "niche anchoring" of LSC affects disease evolution. To address this issue, we conditionally inactivated the adhesion molecule JAM-C (Junctional Adhesion Molecule-C) expressed by hematopoietic stem cells (HSCs) and LSCs in an inducible mixed-lineage leukemia (iMLL)-AF9-driven AML mouse model. Deletion of Jam3 (encoding JAM-C) before induction of the leukemia-initiating iMLL-AF9 fusion resulted in a shift from long-term to short-term HSC expansion, without affecting disease initiation and progression. In vitro experiments showed that JAM-C controlled leukemic cell nesting irrespective of the bone marrow stromal cells used. RNA sequencing performed on leukemic HSCs isolated from diseased mice revealed that genes upregulated in Jam3-deficient animals belonged to activation protein-1 (AP-1) and tumor necrosis factor α (TNF-α)/NF-κB pathways. Human orthologs of dysregulated genes allowed to identify a score that was distinct from, and complementary to, the LSC-17 score. Substratification of patients with AML using LSC-17 and AP-1/TNF-α genes signature defined 4 groups with median survival ranging from <1 year to a median of "not reached" after 8 years. Finally, coculture experiments showed that AP-1 activation in leukemic cells was dependent on the nature of stromal cells. Altogether, our results identify the AP-1/TNF-α gene signature as a proxy of LSC anchoring in bone marrow niches, which improves the prognostic value of the LSC-17 score. This trial was registered at www.ClinicalTrials.gov as #NCT02320656.
Collapse
Affiliation(s)
- Julien M. P. Grenier
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
- UMR 7268, Aix-Marseille Université, EFS, CNRS, GENGLOBE, Marseille, France
| | - Céline Testut
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Matthieu Bal
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Florence Bardin
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Maria De Grandis
- Aix-Marseille University, CNRS, EFS, ADES, Biologie des Groupes Sanguins, Marseille, France
- UMR 7268, Aix-Marseille Université, EFS, CNRS, GENGLOBE, Marseille, France
| | - Véronique Gelsi-Boyer
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Julien Vernerey
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Marjorie Delahaye
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Samuel Granjeaud
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Christophe Zemmour
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Jean-François Spinella
- Laboratory of Molecular Genetics of Stem Cells, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Stéphane J. C. Mancini
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Jean-Marie Boher
- Département de la Recherche Clinique et de l’Innovation, Institut Paoli-Calmettes, Marseille, France
| | - Josée Hébert
- Division of Hematology-Oncology, Department of Medicine, Maisonneuve-Rosemont Hospital, Université de Montréal, Montreal, QC, Canada
| | - Guy Sauvageau
- Laboratory of Molecular Genetics of Stem Cells, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Norbert Vey
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Jürg Schwaller
- Department of Biomedicine, University Children’s Hospital, University of Basel, Basel, Switzerland
| | | | - Cyril Fauriat
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| | - Michel Aurrand-Lions
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Equipe Labellisée Ligue 2020, Marseille, France
| |
Collapse
|
14
|
D'Antonio C, Liguori GL. Dormancy and awakening of cancer cells: the extracellular vesicle-mediated cross-talk between Dr. Jekill and Mr. Hyde. Front Immunol 2024; 15:1441914. [PMID: 39301024 PMCID: PMC11410588 DOI: 10.3389/fimmu.2024.1441914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
Cancer cell dormancy is a reversible process whereby cancer cells enter a quiescent state characterized by cell cycle arrest, inhibition of cell migration and invasion, and increased chemoresistance. Because of its reversibility and resistance to treatment, dormancy is a key process to study, monitor, and interfere with, in order to prevent tumor recurrence and metastasis and improve the prognosis of cancer patients. However, to achieve this goal, further studies are needed to elucidate the mechanisms underlying this complex and dynamic dual process. Here, we review the contribution of extracellular vesicles (EVs) to the regulation of cancer cell dormancy/awakening, focusing on the cross-talk between tumor and non-tumor cells in both the primary tumor and the (pre-)metastatic niche. Although EVs are recognized as key players in tumor progression and metastasis, as well as in tumor diagnostics and therapeutics, their role specifically in dormancy induction/escape is still largely elusive. We report on the most recent and promising results on this topic, focusing on the EV-associated nucleic acids involved. We highlight how EV studies could greatly contribute to the identification of dormancy signaling pathways and a dormancy/early awakening signature for the development of successful diagnostic/prognostic and therapeutic approaches.
Collapse
Affiliation(s)
- Concetta D'Antonio
- Institute of Genetics and Biophysics (IGB) "Adriano Buzzati-Traverso", National Research Council (CNR) of Italy, Naples, Italy
| | - Giovanna L Liguori
- Institute of Genetics and Biophysics (IGB) "Adriano Buzzati-Traverso", National Research Council (CNR) of Italy, Naples, Italy
| |
Collapse
|
15
|
Das N, Panda D, Gajendra S, Gupta R, Thakral D, Kaur G, Khan A, Singh VK, Vemprala A, Bakhshi S, Seth R, Sahoo RK, Sharma A, Rai S, Prajapati VK, Singh S. Immunophenotypic characterization of leukemic stem cells in acute myeloid leukemia using single tube 10-colour panel by multiparametric flow cytometry: Deciphering the spectrum, complexity and immunophenotypic heterogeneity. Int J Lab Hematol 2024; 46:646-656. [PMID: 38456256 DOI: 10.1111/ijlh.14250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024]
Abstract
INTRODUCTION Despite extensive research, comprehensive characterization of leukaemic stem cells (LSC) and information on their immunophenotypic differences from normal haematopoietic stem cells (HSC) is lacking. Herein, we attempted to unravel the immunophenotypic (IPT) characteristics and heterogeneity of LSC using multiparametric flow cytometry (MFC) and single-cell sequencing. MATERIALS AND METHODS Bone marrow aspirate samples from patients with acute myeloid leukaemia (AML) were evaluated using MFC at diagnostic and post induction time points using a single tube-10-colour-panel containing LSC-associated antibodies CD123, CD45RA, CD44, CD33 and COMPOSITE (CLL-1, TIM-3, CD25, CD11b, CD22, CD7, CD56) with backbone markers that is, CD45, CD34, CD38, CD117, sCD3. Single-cell sequencing of the whole transcriptome was also done in a bone marrow sample. RESULTS LSCs and HSCs were identified in 225/255 (88.2%) and 183/255 (71.6%) samples, respectively. Significantly higher expression was noted for COMPOSITE, CD45RA, CD123, CD33, and CD44 in LSCs than HSCs (p < 0.0001). On comparing the LSC specific antigen expressions between CD34+ (n = 184) and CD34- LSCs (n = 41), no difference was observed between the groups. More than one sub-population of LSC was demonstrated in 4.4% of cases, which further revealed high concordance between MFC and single cell transcriptomic analysis in one of the cases displaying three LSC subpopulations by both methods. CONCLUSION A single tube-10-colour MFC panel is proposed as an easy and reproducible tool to identify and discriminate LSCs from HSCs. LSCs display both inter- and intra-sample heterogeneity in terms of antigen expressions, which opens the facets for single cell molecular analysis to elucidate the role of subpopulations of LSCs in AML progression.
Collapse
Affiliation(s)
- Nupur Das
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Devasis Panda
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Smeeta Gajendra
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Ritu Gupta
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Deepshi Thakral
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Gurvinder Kaur
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Aafreen Khan
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Vivek Kumar Singh
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Arushi Vemprala
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Rachna Seth
- Department of Paediatrics, Dr. BRAIRCH, AIIMS, New Delhi, India
| | | | - Atul Sharma
- Department of Medical Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Sandeep Rai
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Vijay K Prajapati
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| | - Saroj Singh
- Department of Laboratory Oncology, Dr. BRAIRCH, AIIMS, New Delhi, India
| |
Collapse
|
16
|
Choi HS, Kim BS, Yoon S, Oh SO, Lee D. Leukemic Stem Cells and Hematological Malignancies. Int J Mol Sci 2024; 25:6639. [PMID: 38928344 PMCID: PMC11203822 DOI: 10.3390/ijms25126639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
The association between leukemic stem cells (LSCs) and leukemia development has been widely established in the context of genetic alterations, epigenetic pathways, and signaling pathway regulation. Hematopoietic stem cells are at the top of the bone marrow hierarchy and can self-renew and progressively generate blood and immune cells. The microenvironment, niche cells, and complex signaling pathways that regulate them acquire genetic mutations and epigenetic alterations due to aging, a chronic inflammatory environment, stress, and cancer, resulting in hematopoietic stem cell dysregulation and the production of abnormal blood and immune cells, leading to hematological malignancies and blood cancer. Cells that acquire these mutations grow at a faster rate than other cells and induce clone expansion. Excessive growth leads to the development of blood cancers. Standard therapy targets blast cells, which proliferate rapidly; however, LSCs that can induce disease recurrence remain after treatment, leading to recurrence and poor prognosis. To overcome these limitations, researchers have focused on the characteristics and signaling systems of LSCs and therapies that target them to block LSCs. This review aims to provide a comprehensive understanding of the types of hematopoietic malignancies, the characteristics of leukemic stem cells that cause them, the mechanisms by which these cells acquire chemotherapy resistance, and the therapies targeting these mechanisms.
Collapse
Affiliation(s)
- Hee-Seon Choi
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
17
|
Wang X, Zhong F, Chen T, Wang H, Wang W, Jin H, Li C, Guo X, Liu Y, Zhang Y, Li B. Cholesterol neutralized vemurafenib treatment by promoting melanoma stem-like cells via its metabolite 27-hydroxycholesterol. Cell Mol Life Sci 2024; 81:226. [PMID: 38775844 PMCID: PMC11111659 DOI: 10.1007/s00018-024-05267-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/14/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Vemurafenib has been used as first-line therapy for unresectable or metastatic melanoma with BRAFV600E mutation. However, overall survival is still limited due to treatment resistance after about one year. Therefore, identifying new therapeutic targets for melanoma is crucial for improving clinical outcomes. In the present study, we found that lowering intracellular cholesterol by knocking down DHCR24, the limiting synthetase, impaired tumor cell proliferation and migration and abrogated the ability to xenotransplant tumors. More importantly, administration of DHCR24 or cholesterol mediated resistance to vemurafenib and promoted the growth of melanoma spheroids. Mechanistically, we identified that 27-hydroxycholesterol (27HC), a primary metabolite of cholesterol synthesized by the enzyme cytochrome P450 27A1 (CYP27A1), reproduces the phenotypes induced by DHCR24 or cholesterol administration and activates Rap1-PI3K/AKT signaling. Accordingly, CYP27A1 is highly expressed in melanoma patients and upregulated by DHCR24 induction. Dafadine-A, a CYP27A1 inhibitor, attenuates cholesterol-induced growth of melanoma spheroids and abrogates the resistance property of vemurafenib-resistant melanoma cells. Finally, we confirmed that the effects of cholesterol on melanoma resistance require its metabolite 27HC through CYP27A1 catalysis, and that 27HC further upregulates Rap1A/Rap1B expression and increases AKT phosphorylation. Thus, our results suggest that targeting 27HC may be a useful strategy to overcome treatment resistance in metastatic melanoma.
Collapse
Affiliation(s)
- Xiaohong Wang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Feiliang Zhong
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, People's Republic of China
| | - Tingting Chen
- School of Basic Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Hongbo Wang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Weifang Wang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Hongkai Jin
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Chouyang Li
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Xuan Guo
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Ying Liu
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Yu Zhang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| | - Bo Li
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-Related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| |
Collapse
|
18
|
de Jong MME, Chen L, Raaijmakers MHGP, Cupedo T. Bone marrow inflammation in haematological malignancies. Nat Rev Immunol 2024:10.1038/s41577-024-01003-x. [PMID: 38491073 DOI: 10.1038/s41577-024-01003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 03/18/2024]
Abstract
Tissue inflammation is a hallmark of tumour microenvironments. In the bone marrow, tumour-associated inflammation impacts normal niches for haematopoietic progenitor cells and mature immune cells and supports the outgrowth and survival of malignant cells residing in these niche compartments. This Review provides an overview of our current understanding of inflammatory changes in the bone marrow microenvironment of myeloid and lymphoid malignancies, using acute myeloid leukaemia and multiple myeloma as examples and highlights unique and shared features of inflammation in niches for progenitor cells and plasma cells. Importantly, inflammation exerts profoundly different effects on normal bone marrow niches in these malignancies, and we provide context for possible drivers of these divergent effects. We explore the role of tumour cells in inflammatory changes, as well as the role of cellular constituents of normal bone marrow niches, including myeloid cells and stromal cells. Integrating knowledge of disease-specific dynamics of malignancy-associated bone marrow inflammation will provide a necessary framework for future targeting of these processes to improve patient outcome.
Collapse
Affiliation(s)
- Madelon M E de Jong
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Lanpeng Chen
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Tom Cupedo
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
19
|
Zarou MM, Rattigan KM, Sarnello D, Shokry E, Dawson A, Ianniciello A, Dunn K, Copland M, Sumpton D, Vazquez A, Helgason GV. Inhibition of mitochondrial folate metabolism drives differentiation through mTORC1 mediated purine sensing. Nat Commun 2024; 15:1931. [PMID: 38431691 PMCID: PMC10908830 DOI: 10.1038/s41467-024-46114-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/07/2024] [Indexed: 03/05/2024] Open
Abstract
Supporting cell proliferation through nucleotide biosynthesis is an essential requirement for cancer cells. Hence, inhibition of folate-mediated one carbon (1C) metabolism, which is required for nucleotide synthesis, has been successfully exploited in anti-cancer therapy. Here, we reveal that mitochondrial folate metabolism is upregulated in patient-derived leukaemic stem cells (LSCs). We demonstrate that inhibition of mitochondrial 1C metabolism through impairment of de novo purine synthesis has a cytostatic effect on chronic myeloid leukaemia (CML) cells. Consequently, changes in purine nucleotide levels lead to activation of AMPK signalling and suppression of mTORC1 activity. Notably, suppression of mitochondrial 1C metabolism increases expression of erythroid differentiation markers. Moreover, we find that increased differentiation occurs independently of AMPK signalling and can be reversed through reconstitution of purine levels and reactivation of mTORC1. Of clinical relevance, we identify that combination of 1C metabolism inhibition with imatinib, a frontline treatment for CML patients, decreases the number of therapy-resistant CML LSCs in a patient-derived xenograft model. Our results highlight a role for folate metabolism and purine sensing in stem cell fate decisions and leukaemogenesis.
Collapse
Affiliation(s)
- Martha M Zarou
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Kevin M Rattigan
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Daniele Sarnello
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Engy Shokry
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Amy Dawson
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Angela Ianniciello
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Karen Dunn
- Paul O'Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - Mhairi Copland
- Paul O'Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G12 0ZD, UK
| | - David Sumpton
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Alexei Vazquez
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
| | - G Vignir Helgason
- Wolfson Wohl Cancer Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
20
|
Jin W, Dai Y, Chen L, Zhu H, Dong F, Zhu H, Meng G, Li J, Chen S, Chen Z, Fang H, Wang K. Cellular hierarchy insights reveal leukemic stem-like cells and early death risk in acute promyelocytic leukemia. Nat Commun 2024; 15:1423. [PMID: 38365836 PMCID: PMC10873341 DOI: 10.1038/s41467-024-45737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
Acute promyelocytic leukemia (APL) represents a paradigm for targeted differentiation therapy, with a minority of patients experiencing treatment failure and even early death. We here report a comprehensive single-cell analysis of 16 APL patients, uncovering cellular compositions and their impact on all-trans retinoic acid (ATRA) response in vivo and early death. We unveil a cellular differentiation hierarchy within APL blasts, rooted in leukemic stem-like cells. The oncogenic PML/RARα fusion protein exerts branch-specific regulation in the APL trajectory, including stem-like cells. APL cohort analysis establishes an association of leukemic stemness with elevated white blood cell counts and FLT3-ITD mutations. Furthermore, we construct an APL-specific stemness score, which proves effective in assessing early death risk. Finally, we show that ATRA induces differentiation of primitive blasts and patients with early death exhibit distinct stemness-associated transcriptional programs. Our work provides a thorough survey of APL cellular hierarchies, offering insights into cellular dynamics during targeted therapy.
Collapse
Affiliation(s)
- Wen Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Honghu Zhu
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Fangyi Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongming Zhu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guoyu Meng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Junmin Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Saijuan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Kankan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
21
|
Aoki T, Shiba N, Tsujimoto S, Yamato G, Hara Y, Kato S, Yoshida K, Ogawa S, Hayashi Y, Iwamoto S, Taki T, Shimada A, Iijima-Yamashita Y, Horibe K, Tawa A, Taga T, Adachi S, Tomizawa D. High IL2RA/CD25 expression is a prognostic stem cell biomarker for pediatric acute myeloid leukemia without a core-binding factor. Pediatr Blood Cancer 2024; 71:e30803. [PMID: 38054691 DOI: 10.1002/pbc.30803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/19/2023] [Accepted: 11/25/2023] [Indexed: 12/07/2023]
Abstract
CD25 is an aberrant marker expressed on the leukemic stem cell (LSC) surface and an immunotherapy target in acute myeloid leukemia (AML). However, the clinical prevalence and significance of CD25 expression in pediatric AML are unknown. High IL2RA/CD25 expression in pediatric AML showed a stem cell-like phenotype, and elevated CD25 expression was associated with lower overall survival (p < .001) and event-free survival (p < .001) in the Japanese Pediatric Leukemia/Lymphoma Study Group AML-05 study. This finding was reproduced in AML without a core-binding factor in the Children's Oncology Group study cohort. High CD25 expression has prognostic significance in pediatric AML.
Collapse
Affiliation(s)
- Takahiro Aoki
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Norio Shiba
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shinichi Tsujimoto
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Genki Yamato
- Department of Pediatrics, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yusuke Hara
- Department of Pediatrics, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Shota Kato
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenichi Yoshida
- Division of Cancer Evolution, National Cancer Center Research Institute, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Yasuhide Hayashi
- Institute of Physiology and Medicine, Jobu University, Takasaki, Japan
| | | | - Tomohiko Taki
- Department of Medical Technology, Kyorin University Faculty of Health Sciences, Mitaka, Japan
| | - Akira Shimada
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | | | - Keizo Horibe
- Clinical Research Center, NHO Nagoya Medical Center, Nagoya, Japan
| | - Akio Tawa
- Higashiosaka Aramoto Heiwa Clinic, Higashiosaka, Japan
| | - Takashi Taga
- Department of Pediatrics, Shiga University of Medical Science, Shiga, Japan
| | - Souichi Adachi
- Department of Human Health Science, Kyoto University, Kyoto, Japan
| | - Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
22
|
Allert C, Müller-Tidow C, Blank MF. The relevance of the hematopoietic niche for therapy resistance in acute myeloid leukemia. Int J Cancer 2024; 154:197-209. [PMID: 37565773 DOI: 10.1002/ijc.34684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
The expansion of acute myeloid leukemia (AML) blasts not only suppresses normal hematopoiesis, but also alters the microenvironment. The interplay of different components of the bone marrow gives rise to altered metabolic states and activates signaling pathways which lead to resistance and impede effective therapy. Therefore, the underlying processes and mechanisms represent attractive therapeutic leverage points for overcoming therapy resistance in AML. Here, we briefly discuss resistance mechanisms based on cell interactions and secreted soluble factors in the hematopoietic niche and provide an overview of niche-related therapeutic targets currently undergoing preclinical and clinical investigation which may help improve the outcome in AML therapy.
Collapse
Affiliation(s)
- Catana Allert
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- University of Heidelberg Medical Faculty, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- University of Heidelberg Medical Faculty, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Maximilian Felix Blank
- Department of Medicine V, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), University of Heidelberg and European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
Jagurinoski M, Davidkova Y, Stojcov-Jagurinoska M, Balatzenko G, Spassov B, Guenova M. Secondary acute myeloid leukemia and de novo acute myeloid leukemia with myelodysplasia-related changes - close or complete strangers? Folia Med (Plovdiv) 2023; 65:728-736. [PMID: 38351754 DOI: 10.3897/folmed.65.e98404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/14/2023] [Indexed: 02/16/2024] Open
Abstract
AIM To compare the main features of patients with secondary acute myeloid leukemias (AMLs) after post-myelodysplastic syndrome (AML-post-MDS) or post-myeloproliferative neoplasms (AML-post-MPN) and myeloid blast crisis of chronic myeloid leukemia (CML-BC) vs. de novoAMLs with myelodysplastic characteristics (dn-AML-MDS).
Collapse
Affiliation(s)
- Milan Jagurinoski
- National Specialized Hospital for Active Treatment of Hematological Diseases, Sofia, Bulgaria
| | - Yanitsa Davidkova
- National Specialized Hospital for Active Treatment of Hematological Diseases, Sofia, Bulgaria
| | | | - Gueorgui Balatzenko
- National Specialized Hospital for Active Treatment of Hematological Diseases, Sofia, Bulgaria
| | - Branimir Spassov
- National Specialized Hospital for Active Treatment of Hematological Diseases, Sofia, Bulgaria
| | - Margarita Guenova
- National Specialized Hospital for Active Treatment of Hematological Diseases, Sofia, Bulgaria
| |
Collapse
|
24
|
Xu YF, Wang GY, Zhang MY, Yang JG. Hub genes and their key effects on prognosis of Burkitt lymphoma. World J Clin Oncol 2023; 14:357-372. [PMID: 37970111 PMCID: PMC10631346 DOI: 10.5306/wjco.v14.i10.357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Burkitt lymphoma (BL) is an exceptionally aggressive malignant neoplasm that arises from either the germinal center or post-germinal center B cells. Patients with BL often present with rapid tumor growth and require high-intensity multi-drug therapy combined with adequate intrathecal chemotherapy prophylaxis, however, a standard treatment program for BL has not yet been established. It is important to identify biomarkers for predicting the prognosis of BLs and discriminating patients who might benefit from the therapy. Microarray data and sequencing information from public databases could offer opportunities for the discovery of new diagnostic or therapeutic targets. AIM To identify hub genes and perform gene ontology (GO) and survival analysis in BL. METHODS Gene expression profiles and clinical traits of BL patients were collected from the Gene Expression Omnibus database. Weighted gene co-expression network analysis (WGCNA) was applied to construct gene co-expression modules, and the cytoHubba tool was used to find the hub genes. Then, the hub genes were analyzed using GO and Kyoto Encyclopedia of Genes and Genomes analysis. Additionally, a Protein-Protein Interaction network and a Genetic Interaction network were constructed. Prognostic candidate genes were identified through overall survival analysis. Finally, a nomogram was established to assess the predictive value of hub genes, and drug-gene interactions were also constructed. RESULTS In this study, we obtained 8 modules through WGCNA analysis, and there was a significant correlation between the yellow module and age. Then we identified 10 hub genes (SRC, TLR4, CD40, STAT3, SELL, CXCL10, IL2RA, IL10RA, CCR7 and FCGR2B) by cytoHubba tool. Within these hubs, two genes were found to be associated with OS (CXCL10, P = 0.029 and IL2RA, P = 0.0066) by survival analysis. Additionally, we combined these two hub genes and age to build a nomogram. Moreover, the drugs related to IL2RA and CXCL10 might have a potential therapeutic role in relapsed and refractory BL. CONCLUSION From WGCNA and survival analysis, we identified CXCL10 and IL2RA that might be prognostic markers for BL.
Collapse
Affiliation(s)
- Yan-Feng Xu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Guan-Yun Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ming-Yu Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ji-Gang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
25
|
Liang G, Wang L, You Q, Cahill K, Chen C, Zhang W, Fulton N, Stock W, Odenike O, He C, Han D. Cellular Composition and 5hmC Signature Predict the Treatment Response of AML Patients to Azacitidine Combined with Chemotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300445. [PMID: 37271891 PMCID: PMC10427370 DOI: 10.1002/advs.202300445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/12/2023] [Indexed: 06/06/2023]
Abstract
Azacitidine (AZA) is a DNA methyltransferase inhibitor and epigenetic modulator that can be an effective agent in combination with chemotherapy for patients with high-risk acute myeloid leukemia (AML). However, biological factors driving the therapeutic response of such hypomethylating agent (HMA)-based therapies remain unknown. Herein, the transcriptome and/or genome-wide 5-hydroxymethylcytosine (5hmC) is characterized for 41 patients with high-risk AML from a phase 1 clinical trial treated with AZA epigenetic priming followed by high-dose cytarabine and mitoxantrone (AZA-HiDAC-Mito). Digital cytometry reveals that responders have elevated Granulocyte-macrophage-progenitor-like (GMP-like) malignant cells displaying an active cell cycle program. Moreover, the enrichment of natural killer (NK) cells predicts a favorable outcome in patients receiving AZA-HiDAC-Mito therapy or other AZA-based therapies. Comparing 5hmC profiles before and after five-day treatment of AZA shows that AZA exposure induces dose-dependent 5hmC changes, in which the magnitude correlates with overall survival (p = 0.015). An extreme gradient boosting (XGBoost) machine learning model is developed to predict the treatment response based on 5hmC levels of 11 genes, achieving an area under the curve (AUC) of 0.860. These results suggest that cellular composition markedly impacts the treatment response, and showcase the prospect of 5hmC signatures in predicting the outcomes of HMA-based therapies in AML.
Collapse
Affiliation(s)
- Guanghao Liang
- Key Laboratory of Genomic and Precision MedicineBeijing Institute of GenomicsChinese Academy of Sciences and China National Center for BioinformationBeijing100101China
- College of Future TechnologySino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100049China
| | - Linchen Wang
- Key Laboratory of Genomic and Precision MedicineBeijing Institute of GenomicsChinese Academy of Sciences and China National Center for BioinformationBeijing100101China
- College of Future TechnologySino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100049China
| | - Qiancheng You
- Department of Chemistry and Institute for Biophysical DynamicsThe University of ChicagoChicagoIL60637USA
- Howard Hughes Medical InstituteChicagoIL60637USA
| | - Kirk Cahill
- Section of Hematology/OncologyDepartment of MedicineUniversity of Chicago MedicineChicagoIL60637USA
| | - Chuanyuan Chen
- Key Laboratory of Genomic and Precision MedicineBeijing Institute of GenomicsChinese Academy of Sciences and China National Center for BioinformationBeijing100101China
- College of Future TechnologySino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100049China
| | - Wei Zhang
- Department of MedicineUniversity of California, San DiegoLa JollaCA92093USA
- Bristol‐Myers SquibbSan DiegoCA92121USA
| | - Noreen Fulton
- Section of Hematology/OncologyDepartment of MedicineUniversity of Chicago MedicineChicagoIL60637USA
- Comprehensive Cancer CenterUniversity of Chicago MedicineChicagoIL60637USA
| | - Wendy Stock
- Section of Hematology/OncologyDepartment of MedicineUniversity of Chicago MedicineChicagoIL60637USA
- Comprehensive Cancer CenterUniversity of Chicago MedicineChicagoIL60637USA
| | - Olatoyosi Odenike
- Section of Hematology/OncologyDepartment of MedicineUniversity of Chicago MedicineChicagoIL60637USA
- Comprehensive Cancer CenterUniversity of Chicago MedicineChicagoIL60637USA
| | - Chuan He
- Department of Chemistry and Institute for Biophysical DynamicsThe University of ChicagoChicagoIL60637USA
- Howard Hughes Medical InstituteChicagoIL60637USA
- Department of Biochemistry and Molecular BiologyThe University of ChicagoChicagoIL60637USA
| | - Dali Han
- Key Laboratory of Genomic and Precision MedicineBeijing Institute of GenomicsChinese Academy of Sciences and China National Center for BioinformationBeijing100101China
- College of Future TechnologySino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| |
Collapse
|
26
|
Edwards K, Lydyard PM, Kulikova N, Tsertsvadze T, Volpi EV, Chiorazzi N, Porakishvili N. The role of CD180 in hematological malignancies and inflammatory disorders. Mol Med 2023; 29:97. [PMID: 37460961 PMCID: PMC10353253 DOI: 10.1186/s10020-023-00682-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023] Open
Abstract
Toll-like receptors play a significant role in the innate immune system and are also involved in the pathophysiology of many different diseases. Over the past 35 years, there have been a growing number of publications exploring the role of the orphan toll-like receptor, CD180. We therefore set out to provide a narrative review of the current evidence surrounding CD180 in both health and disease. We first explore the evidence surrounding the role of CD180 in physiology including its expression, function and signaling in antigen presenting cells (APCs) (dendritic cells, monocytes, and B cells). We particularly focus on the role of CD180 as a modulator of other TLRs including TLR2, TLR4, and TLR9. We then discuss the role of CD180 in inflammatory and autoimmune diseases, as well as in hematological malignancies of B cell origin, including chronic lymphocytic leukemia (CLL). Based on this evidence we produce a current model for CD180 in disease and explore the potential role for CD180 as both a prognostic biomarker and therapeutic target. Throughout, we highlight specific areas of research which should be addressed to further the understanding of CD180 biology and the translational potential of research into CD180 in various diseases.
Collapse
Affiliation(s)
- Kurtis Edwards
- School of Life Sciences, University of Westminster, London, UK
| | - Peter M Lydyard
- School of Life Sciences, University of Westminster, London, UK.
- The University of Georgia, Tbilisi, Georgia.
- Division of Infection of Immunity, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Nino Kulikova
- Agricultural University of Georgia, Tbilisi, Georgia
| | | | | | | | | |
Collapse
|
27
|
Liang XW, Liu B, Chen JC, Cao Z, Chu FR, Lin X, Wang SZ, Wu JC. Characteristics and molecular mechanism of drug-tolerant cells in cancer: a review. Front Oncol 2023; 13:1177466. [PMID: 37483492 PMCID: PMC10360399 DOI: 10.3389/fonc.2023.1177466] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Drug resistance in tumours has seriously hindered the therapeutic effect. Tumour drug resistance is divided into primary resistance and acquired resistance, and the recent study has found that a significant proportion of cancer cells can acquire stable drug resistance from scratch. This group of cells first enters the drug tolerance state (DT state) under drug pressure, and gradually acquires stable drug resistance through adaptive mutations in this state. Although the specific mechanisms underlying the formation of drug tolerant cells (DTCs) remain unclear, various proteins and signalling pathways have been identified as being involved in the formation of DTCs. In the current review, we summarize the characteristics, molecular mechanisms and therapeutic strategies of DTCs in detail.
Collapse
Affiliation(s)
- Xian-Wen Liang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Bing- Liu
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Jia-Cheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Zhi Cao
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Feng-ran Chu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Xiong Lin
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Sheng-Zhong Wang
- Department of Gastrointestinal Surgery, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Jin-Cai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| |
Collapse
|
28
|
Shao R, Li Z, Xin H, Jiang S, Zhu Y, Liu J, Huang R, Xu K, Shi X. Biomarkers as targets for CAR-T/NK cell therapy in AML. Biomark Res 2023; 11:65. [PMID: 37330575 PMCID: PMC10276424 DOI: 10.1186/s40364-023-00501-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/11/2023] [Indexed: 06/19/2023] Open
Abstract
The most common kind of acute leukemia in adults is acute myeloid leukemia (AML), which is often treated with induction chemotherapy regimens followed by consolidation or allogeneic hematopoietic stem cell transplantation (HSCT). However, some patients continue to develop relapsed or refractory AML (R/R-AML). Small molecular targeted drugs require long-time administration. Not all the patients hold molecular targets. Novel medicines are therefore needed to enhance treatment outcomes. T cells and natural killer (NK) cells engineered with chimeric antigen receptors (CARs) that target antigens associated with AML have recently been produced and are currently being tested in both pre-clinical and clinical settings. This review provides an overview of CAR-T/NK treatments for AML.
Collapse
Affiliation(s)
- Ruonan Shao
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Zijian Li
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Honglei Xin
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Suyu Jiang
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Yilin Zhu
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Jingan Liu
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Rong Huang
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Kailin Xu
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Xiaofeng Shi
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China.
| |
Collapse
|
29
|
Luo Q, Liu P, Yu P, Qin T. Cancer Stem Cells are Actually Stem Cells with Disordered Differentiation: the Monophyletic Origin of Cancer. Stem Cell Rev Rep 2023; 19:827-838. [PMID: 36648606 PMCID: PMC10185654 DOI: 10.1007/s12015-023-10508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
Cancer stem cells (CSCs) play an important role in cancer development. Based on advancements in CSC research, we propose a monophyletic model of cancer. This model is based on the idea that CSCs are stem cells with disordered differentiation whose original purpose was to repair damaged tissues. Inflammatory responses and damage repair signals are crucial for the creation and maintenance of CSCs. Normal quiescent stem cells are activated by environmental stimulation, such as an inflammatory response, and undergo cell division and differentiation. In the initial stage of cancer development, stem cell differentiation leads to heteromorphism due to the accumulation of gene mutations, resulting in the development of metaplasia or precancerosis. In the second stage, accumulated mutations induce poor differentiation and lead to cancer development. The monophyletic model illustrates the evolution, biological behavior, and hallmarks of CSCs, proposes a concise understanding of the origin of cancer, and may encourage a novel therapeutic approach.
Collapse
Affiliation(s)
- Qiankun Luo
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Pan Liu
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Pengfei Yu
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China
| | - Tao Qin
- Department of Hepatobilliary and Pancreatic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Jinshui District, No. 7, Weiwu Rd., Zhengzhou, 450003, Henan, China.
| |
Collapse
|
30
|
Ivanov D, Milosevic Feenstra JD, Sadovnik I, Herrmann H, Peter B, Willmann M, Greiner G, Slavnitsch K, Hadzijusufovic E, Rülicke T, Dahlhoff M, Hoermann G, Machherndl‐Spandl S, Eisenwort G, Fillitz M, Sliwa T, Krauth M, Bettelheim P, Sperr WR, Koller E, Pfeilstöcker M, Gisslinger H, Keil F, Kralovics R, Valent P. Phenotypic characterization of disease-initiating stem cells in JAK2- or CALR-mutated myeloproliferative neoplasms. Am J Hematol 2023; 98:770-783. [PMID: 36814396 PMCID: PMC10952374 DOI: 10.1002/ajh.26889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/07/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023]
Abstract
Myeloproliferative neoplasms (MPN) are characterized by uncontrolled expansion of myeloid cells, disease-related mutations in certain driver-genes including JAK2, CALR, and MPL, and a substantial risk to progress to secondary acute myeloid leukemia (sAML). Although behaving as stem cell neoplasms, little is known about disease-initiating stem cells in MPN. We established the phenotype of putative CD34+ /CD38- stem cells and CD34+ /CD38+ progenitor cells in MPN. A total of 111 patients with MPN suffering from polycythemia vera, essential thrombocythemia, or primary myelofibrosis (PMF) were examined. In almost all patients tested, CD34+ /CD38- stem cells expressed CD33, CD44, CD47, CD52, CD97, CD99, CD105, CD117, CD123, CD133, CD184, CD243, and CD274 (PD-L1). In patients with PMF, MPN stem cells often expressed CD25 and sometimes also CD26 in an aberrant manner. MPN stem cells did not exhibit substantial amounts of CD90, CD273 (PD-L2), CD279 (PD-1), CD366 (TIM-3), CD371 (CLL-1), or IL-1RAP. The phenotype of CD34+ /CD38- stem cells did not change profoundly during progression to sAML. The disease-initiating capacity of putative MPN stem cells was confirmed in NSGS mice. Whereas CD34+ /CD38- MPN cells engrafted in NSGS mice, no substantial engraftment was produced by CD34+ /CD38+ or CD34- cells. The JAK2-targeting drug fedratinib and the BRD4 degrader dBET6 induced apoptosis and suppressed proliferation in MPN stem cells. Together, MPN stem cells display a unique phenotype, including cytokine receptors, immune checkpoint molecules, and other clinically relevant target antigens. Phenotypic characterization of neoplastic stem cells in MPN and sAML should facilitate their enrichment and the development of stem cell-eradicating (curative) therapies.
Collapse
Affiliation(s)
- Daniel Ivanov
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | | | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | - Harald Herrmann
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department of Radiation OncologyMedical University of ViennaViennaAustria
| | - Barbara Peter
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | - Michael Willmann
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department for Companion Animals, Clinical Unit for Internal MedicineUniversity of Veterinary Medicine ViennaViennaAustria
| | - Georg Greiner
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
- Ihr Labor, Medical Diagnostic LaboratoriesViennaAustria
| | - Katharina Slavnitsch
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Emir Hadzijusufovic
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department for Companion Animals, Clinical Unit for Internal MedicineUniversity of Veterinary Medicine ViennaViennaAustria
| | - Thomas Rülicke
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Department of Biomedical SciencesUniversity of Veterinary Medicine ViennaViennaAustria
| | - Maik Dahlhoff
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Institute of in vivo and in vitro ModelsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- MLL Munich Leukemia LaboratoryMunichGermany
| | - Sigrid Machherndl‐Spandl
- Hospital Ordensklinikum Elisabethinen LinzLinzAustria
- Johannes Kepler University, Medical FacultyLinzAustria
| | - Gregor Eisenwort
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Michael Fillitz
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Thamer Sliwa
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Maria‐Theresa Krauth
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | | | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| | - Elisabeth Koller
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Michael Pfeilstöcker
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Heinz Gisslinger
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
| | - Felix Keil
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
- Third Medical Department for Hematology and OncologyHanusch Hospital ViennaViennaAustria
| | - Robert Kralovics
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology and OncologyMedical University of ViennaViennaAustria
| |
Collapse
|
31
|
Luo S, Du S, Tao M, Cao J, Cheng P. Insights on hematopoietic cell kinase: An oncogenic player in human cancer. Biomed Pharmacother 2023; 160:114339. [PMID: 36736283 DOI: 10.1016/j.biopha.2023.114339] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Hematopoietic cell kinase (Hck) is a member of the Src family and is expressed in hematopoietic cells. By regulating multiple signaling pathways, HCK can interact with multiple receptors to regulate signaling events involved in cell adhesion, proliferation, migration, invasion, apoptosis, and angiogenesis. However, aberrant expression of Hck in various hematopoietic cells and solid tumors plays a crucial role in tumor-related properties, including cell proliferation and epithelial-mesenchymal transition. In addition, Hck signaling regulates the function of immune cells such as macrophages, contributing to an immunosuppressive tumor microenvironment. The clinical success of various kinase inhibitors targeting the Src kinase family has validated the efficacy of targeting Src, and therapies with highly selective Hck kinase inhibitors are in clinical trials. This article reviews Hck inhibition as an emerging cancer treatment strategy, focusing on the expressions and functions of Hck in tumors and its impact on the tumor microenvironment. It also explores preclinical and clinical pharmacological strategies for Hck targeting to shed light on Hck-targeted tumor therapy.
Collapse
Affiliation(s)
- Shuyan Luo
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Shaonan Du
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Mei Tao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, 300060 Tianjin, China
| | - Jingyuan Cao
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
32
|
Chamo M, Koren O, Goldstein O, Bujanover N, Keinan N, Scharff Y, Gazit R. Molecular Mechanisms in Murine Syngeneic Leukemia Stem Cells. Cancers (Basel) 2023; 15:cancers15030720. [PMID: 36765677 PMCID: PMC9913241 DOI: 10.3390/cancers15030720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is a severe disease with a very high relapse rate. AML relapse may be attributable to leukemic stem cells (LSC). Notably, the "cancer stem cell" theory, which relates to LSCs, is controversial and criticized due to the technical peculiarities of the xenotransplant of human cells into mice. In this study, we searched for possible LSCs in an immunocompetent synergetic mice model. First, we found phenotypic heterogeneity in the ML23 leukemia line. We prospectively isolated a sub-population using the surface markers cKit+CD9-CD48+Mac1-/low, which have the potency to relapse the disease. Importantly, this sub-population can pass in syngeneic hosts and retrieve the heterogeneity of the parental ML23 leukemia line. The LSC sub-population resides in various organs. We present a unique gene expression signature of the LSC in the ML23 model compared to the other sub-populations. Interestingly, the ML23 LSC sub-population expresses therapeutic targeted genes such as CD47 and CD93. Taken together, we present the identification and molecular characterization of LSCs in a syngeneic murine model.
Collapse
|
33
|
Saultz JN, Tyner JW. Chasing leukemia differentiation through induction therapy, relapse and transplantation. Blood Rev 2023; 57:101000. [PMID: 36041918 DOI: 10.1016/j.blre.2022.101000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/23/2022] [Accepted: 08/09/2022] [Indexed: 01/28/2023]
Abstract
Despite rapid advances in our understanding of acute myeloid leukemia (AML), the disease remains challenging to treat with 5-year survival for adult patients 20 years or older estimated to be 26% (Cancer 2021). The use of new targeted therapies including BCL2, IDH1/IDH2, and FLT3 inhibitors has revolutionized treatment approaches but also changed the disease trajectory with unique modes of resistance. Recent studies have shown that stem cell maturation state drives expression level and/or dependence on various pathways, critical to determining drug response. Instead of anticipating these changes, we remain behind the curve chasing the next expanded clone. This review will focus on current approaches to treatment in AML, including defining the significance of blast differentiation state on chemotherapeutic response, signaling pathway dependence, metabolism, immune response, and phenotypic changes. We conclude that multimodal treatment approaches are necessary to target both the immature and mature clones, thereby, sustaining drug response.
Collapse
Affiliation(s)
- Jennifer N Saultz
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States of America; Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, United States of America.
| | - Jeffrey W Tyner
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States of America; Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, United States of America; Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, United States of America
| |
Collapse
|
34
|
Wood CR, Wu WT, Yang YS, Yang JS, Xi Y, Yang WJ. From ecology to oncology: To understand cancer stem cell dormancy, ask a Brine shrimp (Artemia). Adv Cancer Res 2023; 158:199-231. [PMID: 36990533 DOI: 10.1016/bs.acr.2022.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The brine shrimp (Artemia), releases embryos that can remain dormant for up to a decade. Molecular and cellular level controlling factors of dormancy in Artemia are now being recognized or applied as active controllers of dormancy (quiescence) in cancers. Most notably, the epigenetic regulation by SET domain-containing protein 4 (SETD4), is revealed as highly conserved and the primary control factor governing the maintenance of cellular dormancy from Artemia embryonic cells to cancer stem cells (CSCs). Conversely, DEK, has recently emerged as the primary factor in the control of dormancy exit/reactivation, in both cases. The latter has been now successfully applied to the reactivation of quiescent CSCs, negating their resistance to therapy and leading to their subsequent destruction in mouse models of breast cancer, without recurrence or metastasis potential. In this review, we introduce the many mechanisms of dormancy from Artemia ecology that have been translated into cancer biology, and herald Artemia's arrival on the model organism stage. We show how Artemia studies have unlocked the mechanisms of the maintenance and termination of cellular dormancy. We then discuss how the antagonistic balance of SETD4 and DEK fundamentally controls chromatin structure and consequently governs CSCs function, chemo/radiotherapy resistance, and dormancy in cancers. Many key stages from transcription factors to small RNAs, tRNA trafficking, molecular chaperones, ion channels, and links with various pathways and aspects of signaling are also noted, all of which link studies in Artemia to those of cancer on a molecular and/or cellular level. We particularly emphasize that the application of such emerging factors as SETD4 and DEK may open new and clear avenues for the treatment for various human cancers.
Collapse
Affiliation(s)
- Christopher R Wood
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Wen-Tao Wu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yao-Shun Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jin-Shu Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yongmei Xi
- The Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Wei-Jun Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
35
|
Present and Future Role of Immune Targets in Acute Myeloid Leukemia. Cancers (Basel) 2022; 15:cancers15010253. [PMID: 36612249 PMCID: PMC9818182 DOI: 10.3390/cancers15010253] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
It is now well known that the bone marrow (BM) cell niche contributes to leukemogenesis, but emerging data support the role of the complex crosstalk between AML cells and the BM microenvironment to induce a permissive immune setting that protects leukemic stem cells (LSCs) from therapy-induced death, thus favoring disease persistence and eventual relapse. The identification of potential immune targets on AML cells and the modulation of the BM environment could lead to enhanced anti-leukemic effects of drugs, immune system reactivation, and the restoration of AML surveillance. Potential targets and effectors of this immune-based therapy could be monoclonal antibodies directed against LSC antigens such as CD33, CD123, and CLL-1 (either as direct targets or via several bispecific T-cell engagers), immune checkpoint inhibitors acting on different co-inhibitory axes (alone or in combination with conventional AML drugs), and novel cellular therapies such as chimeric antigen receptor (CAR) T-cells designed against AML-specific antigens. Though dozens of clinical trials, mostly in phases I and II, are ongoing worldwide, results have still been negatively affected by difficulties in the identification of the optimal targets on LSCs.
Collapse
|
36
|
Ganzel C, Sun Z, Baslan T, Zhang Y, Gönen M, Abdel-Wahab OI, Racevskis J, Garrett-Bakelman F, Lowe SW, Fernandez HF, Ketterling R, Luger SM, Litzow M, Lazarus HM, Rowe JM, Tallman MS, Levine RL, Paietta E. Measurable residual disease by flow cytometry in acute myeloid leukemia is prognostic, independent of genomic profiling. Leuk Res 2022; 123:106971. [PMID: 36332294 PMCID: PMC9789386 DOI: 10.1016/j.leukres.2022.106971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/04/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
Abstract
Measurable residual disease (MRD) assessment provides a potent indicator of the efficacy of anti-leukemic therapy. It is unknown, however, whether integrating MRD with molecular profiling better identifies patients at risk of relapse. To investigate the clinical relevance of MRD in relation to a molecular-based prognostic schema, we measured MRD by flow cytometry in 189 AML patients enrolled in ECOG-ACRIN E1900 trial (NCT00049517) in morphologic complete remission (CR) (28.8 % of the original cohort) representing 44.4 % of CR patients. MRD positivity was defined as ≥ 0.1 % of leukemic bone marrow cells. Risk classification was based on standard cytogenetics, fluorescence-in-situ-hybridization, somatic gene analysis, and sparse whole genome sequencing for copy number ascertainment. At 84.6 months median follow-up of patients still alive at the time of analysis (range 47.0-120 months), multivariate analysis demonstrated that MRD status at CR (p = 0.001) and integrated molecular risk (p = 0.0004) independently predicted overall survival (OS). Among risk classes, MRD status significantly affected OS only in the favorable risk group (p = 0.002). Expression of CD25 (α-chain of the interleukin-2 receptor) by leukemic myeloblasts at diagnosis negatively affected OS independent of post-treatment MRD levels. These data suggest that integrating MRD with genetic profiling and pre-treatment CD25 expression may improve prognostication in AML.
Collapse
Affiliation(s)
- Chezi Ganzel
- Hematology Department, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| | - Zhuoxin Sun
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar I Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Janis Racevskis
- Department of Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Francine Garrett-Bakelman
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Departments of Medicine and Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, USA; University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hugo F Fernandez
- Malignant Hematology and Cellular Therapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Rhett Ketterling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Selina M Luger
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Litzow
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Jacob M Rowe
- Hematology Department, Shaare Zedek Medical Center, and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Martin S Tallman
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
37
|
Donia HM, Elsweify NM, Farahat NM, Nadwan EA. Wilms tumor 1 gene expression in acute myeloid leukemia: prognostic significance and usefulness in minimal residual disease monitoring—a case–control study. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00291-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Minimal residual disease (MRD), which is characterized as leukemic cells at a level below morphologic detection, has been connected to the risk of relapse in acute myeloid leukemia. In 80–90% of acute myeloid leukemia (AML) patients, the Wilms tumor (WT1) gene is overexpressed at the mRNA level. In our prospective study, a total of 55 patients were enrolled in the study. Group I involved 40 AML patients and group II involved 15 patients healthy controls. WT1 gene expression was quantified using quantitative real-time PCR on bone marrow samples from AML patients at initial diagnosis and at day 28 after induction chemotherapy, and compared to 15 healthy controls in group II. Follow up of patients for prognosis evaluation was assessed. IBM SPSS software was used to capture and analyses the data.
Results
At diagnosis, the mean WT1 transcript value in AML patients was substantially higher than the expression observed in control patient’s Bone marrow. There was no statistically relevant relationship between the onset of relapse and WT1 expression. Patients with WT1 overexpression at diagnosis had a shorter overall survival than patients with negative WT1 expression.
Conclusions
Wilms tumor 1 gene expression was found to be significantly higher in AML patients than control cases, overall, our results confirmed the prognostic significance of WT1 overexpression in AML patients. Our findings support the application of MRD in AML patients based on WT1 overexpression.
Collapse
|
38
|
Role of PARP Inhibitors in Cancer Immunotherapy: Potential Friends to Immune Activating Molecules and Foes to Immune Checkpoints. Cancers (Basel) 2022; 14:cancers14225633. [PMID: 36428727 PMCID: PMC9688455 DOI: 10.3390/cancers14225633] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/04/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) induce cytotoxic effects as single agents in tumors characterized by defective repair of DNA double-strand breaks deriving from BRCA1/2 mutations or other abnormalities in genes associated with homologous recombination. Preclinical studies have shown that PARPi-induced DNA damage may affect the tumor immune microenvironment and immune-mediated anti-tumor response through several mechanisms. In particular, increased DNA damage has been shown to induce the activation of type I interferon pathway and up-regulation of PD-L1 expression in cancer cells, which can both enhance sensitivity to Immune Checkpoint Inhibitors (ICIs). Despite the recent approval of ICIs for a number of advanced cancer types based on their ability to reinvigorate T-cell-mediated antitumor immune responses, a consistent percentage of treated patients fail to respond, strongly encouraging the identification of combination therapies to overcome resistance. In the present review, we analyzed both established and unexplored mechanisms that may be elicited by PARPi, supporting immune reactivation and their potential synergism with currently used ICIs. This analysis may indicate novel and possibly patient-specific immune features that might represent new pharmacological targets of PARPi, potentially leading to the identification of predictive biomarkers of response to their combination with ICIs.
Collapse
|
39
|
Jiang Y, Liu L, Jiang Y, Li Z, Feng L, Zhuang X, Lin Z, Chen Q, Chen G, He J, Li G, Zha J, Xu B. Preclinical Evaluation of the Multiple Tyrosine Kinases Inhibitor Anlotinib in Leukemia Stem Cells. Pharmaceuticals (Basel) 2022; 15:1313. [PMID: 36355485 PMCID: PMC9697152 DOI: 10.3390/ph15111313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/08/2022] [Accepted: 10/21/2022] [Indexed: 10/05/2023] Open
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.
Collapse
Affiliation(s)
- Yuelong Jiang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| | - Long Liu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| | - Yirong Jiang
- Department of Hematology, Affiliated Dongguan People’s Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523059, China
| | - Zhifeng Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| | - Liying Feng
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| | - Xinguo Zhuang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| | - Zhijuan Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| | - Qiuling Chen
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| | - Guoshu Chen
- Department of Hematology, Huizhou Municipal Central Hospital, Huizhou 516001, China
| | - Jixiang He
- Department of Hematology, Affiliated Dongguan People’s Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523059, China
| | - Guowei Li
- Department of Hematology, Huizhou Municipal Central Hospital, Huizhou 516001, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Diagnosis and Therapy for Hematological Malignancies, Xiamen 361003, China
| |
Collapse
|
40
|
Nam AS, Dusaj N, Izzo F, Murali R, Myers RM, Mouhieddine TH, Sotelo J, Benbarche S, Waarts M, Gaiti F, Tahri S, Levine R, Abdel-Wahab O, Godley LA, Chaligne R, Ghobrial I, Landau DA. Single-cell multi-omics of human clonal hematopoiesis reveals that DNMT3A R882 mutations perturb early progenitor states through selective hypomethylation. Nat Genet 2022; 54:1514-1526. [PMID: 36138229 PMCID: PMC10068894 DOI: 10.1038/s41588-022-01179-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 07/29/2022] [Indexed: 12/13/2022]
Abstract
Somatic mutations in cancer genes have been detected in clonal expansions across healthy human tissue, including in clonal hematopoiesis. However, because mutated and wild-type cells are admixed, we have limited ability to link genotypes with phenotypes. To overcome this limitation, we leveraged multi-modality single-cell sequencing, capturing genotype, transcriptomes and methylomes in progenitors from individuals with DNMT3A R882 mutated clonal hematopoiesis. DNMT3A mutations result in myeloid over lymphoid bias, and an expansion of immature myeloid progenitors primed toward megakaryocytic-erythroid fate, with dysregulated expression of lineage and leukemia stem cell markers. Mutated DNMT3A leads to preferential hypomethylation of polycomb repressive complex 2 targets and a specific CpG flanking motif. Notably, the hypomethylation motif is enriched in binding motifs of key hematopoietic transcription factors, serving as a potential mechanistic link between DNMT3A mutations and aberrant transcriptional phenotypes. Thus, single-cell multi-omics paves the road to defining the downstream consequences of mutations that drive clonal mosaicism.
Collapse
Affiliation(s)
- Anna S Nam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Neville Dusaj
- New York Genome Center, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, Rockefeller University, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Franco Izzo
- New York Genome Center, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Rekha Murali
- New York Genome Center, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Robert M Myers
- New York Genome Center, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, Rockefeller University, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tarek H Mouhieddine
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jesus Sotelo
- New York Genome Center, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Salima Benbarche
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Waarts
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Federico Gaiti
- New York Genome Center, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Sabrin Tahri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ross Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lucy A Godley
- Section of Hematology/Oncology, Departments of Medicine and Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Ronan Chaligne
- New York Genome Center, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Irene Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Dan A Landau
- New York Genome Center, New York, NY, USA.
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
41
|
Pingul BY, Huang H, Chen Q, Alikarami F, Zhang Z, Qi J, Bernt KM, Berger SL, Cao Z, Shi J. Dissection of the MEF2D-IRF8 transcriptional circuit dependency in acute myeloid leukemia. iScience 2022; 25:105139. [PMID: 36193052 PMCID: PMC9526175 DOI: 10.1016/j.isci.2022.105139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 08/05/2022] [Accepted: 09/10/2022] [Indexed: 11/26/2022] Open
Abstract
Transcriptional dysregulation is a prominent feature in leukemia. Here, we systematically surveyed transcription factor (TF) vulnerabilities in leukemia and uncovered TF clusters that exhibit context-specific vulnerabilities within and between different subtypes of leukemia. Among these TF clusters, we demonstrated that acute myeloid leukemia (AML) with high IRF8 expression was addicted to MEF2D. MEF2D and IRF8 form an autoregulatory loop via direct binding to mutual enhancer elements. One important function of this circuit in AML is to sustain PU.1/MEIS1 co-regulated transcriptional outputs via stabilizing PU.1’s chromatin occupancy. We illustrated that AML could acquire dependency on this circuit through various oncogenic mechanisms that results in the activation of their enhancers. In addition to forming a circuit, MEF2D and IRF8 can also separately regulate gene expression, and dual perturbation of these two TFs leads to a more robust inhibition of AML proliferation. Collectively, our results revealed a TF circuit essential for AML survival. MEF2D is a context-specific vulnerability in IRF8hi AML MEF2D and IRF8 form a transcriptional circuit via binding to each other’s enhancers MEF2D-IRF8 circuit supports PU.1’s chromatin occupancy and transcriptional output MEF2D and IRF8 can regulate separate gene expression programs alongside the circuit
Collapse
|
42
|
Jia J, Liu B, Wang D, Wang X, Song L, Ren Y, Guo Z, Ma K, Cui C. CD93 promotes acute myeloid leukemia development and is a potential therapeutic target. Exp Cell Res 2022; 420:113361. [PMID: 36152731 DOI: 10.1016/j.yexcr.2022.113361] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/26/2022]
Abstract
CD93 is a transmembrane receptor belonging to the Group XIV C-Type lectin family. It is expressed in a variety of cellular types such as monocytes, neutrophils, platelets, microglia, and endothelial cells. CD93 has been reported to play important roles in cell proliferation, cell migration, and tumor angiogenesis. Here, we show CD93 is highly expressed in M4 and M5 subtypes of acute myeloid leukemia (AML) patients, and highly expressed in leukemia stem cells, AML progenitor cells, as well as more differentiated AML cells. We found that CD93 promotes AML cell proliferation, while CD93 deficient AML cells commit to differentiation. We further show that CD93 exerts its proliferative function through downstream SHP-2/Syk/CREB cascade in AML cells. Moreover, human AML cells treated with CD93 mAb combined with αMFc-NC-DM1 (an IgG Fc specific antibody conjugated to maytansinoid DM1), showed a striking reduction of proliferation. Our study revealed that CD93 is a critical participator of AML development and provides a potential therapeutic cell surface target. (160 words).
Collapse
Affiliation(s)
- Jie Jia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Bin Liu
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Dandan Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Xiaohong Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Lingrui Song
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Yanzhang Ren
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Zhaoming Guo
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Kun Ma
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China
| | - Changhao Cui
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 124221, China.
| |
Collapse
|
43
|
Weeda V, Mestrum SGC, Leers MPG. Flow Cytometric Identification of Hematopoietic and Leukemic Blast Cells for Tailored Clinical Follow-Up of Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:ijms231810529. [PMID: 36142442 PMCID: PMC9506284 DOI: 10.3390/ijms231810529] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML) is a myeloid malignancy that is characterized by the accumulation of leukemic blast cells, which originate from hematopoietic stem cells that have undergone leukemic transformation and/or are more mature progenitors that have gained stemness features. Currently, no consensus exists for the flow cytometric identification of normal blast cells and their leukemic counterparts by their antigenic expression profile. Differentiating between the benign cells and the malignant cells is crucial for the further deployment of immunophenotype panels for the clinical follow-up of AML patients. This review provides an overview of immunophenotypic markers that allow the identification of leukemic blast cells in the bone marrow with multiparameter flow cytometry. This technique allows the identification of hematopoietic blast cells at the level of maturing cells by their antigen expression profile. While aberrant antigen expression of a single immunophenotypic marker cell cannot be utilized in order to differentiate leukemic blast cells from normal blast cells, combinations of multiple immunophenotypic markers can enable the distinction of normal and leukemic blast cells. The identification of these markers has provided new perspectives for tailored clinical follow-up, including therapy management, diagnostics, and prognostic purposes. The immunophenotypic marker panels, however, should be developed by carefully considering the variable antigen marker expression profile of individual patients.
Collapse
Affiliation(s)
- Vera Weeda
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
| | - Stefan G. C. Mestrum
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
- Department of Molecular Cell Biology, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, 6200MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-6-36176124
| | - Math P. G. Leers
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
| |
Collapse
|
44
|
Nakase K, Tanaka I, Tawara I, Shiku H. CD22 expression in acute myeloid leukemia: close correlation with interleukin-2 receptor α-chain (CD25) expression and poor prognosis. Leuk Lymphoma 2022; 63:2251-2253. [PMID: 35442136 DOI: 10.1080/10428194.2022.2062346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Kazunori Nakase
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Hematology and Oncology, Mie University Hospital, Tsu, Japan
| | - Isao Tanaka
- Department of Internal Medicine, Suzuka Kaisei Hospital, Suzuka, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Hospital, Tsu, Japan
| | - Hiroshi Shiku
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
45
|
Tumor cell dormancy: Molecular mechanisms, and pharmacological approaches to target dormant cells for countering tumor. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
46
|
Díaz-Beyá M, García-Fortes M, Valls R, Artigas L, Gómez-Casares MT, Montesinos P, Sánchez-Guijo F, Coma M, Vendranes M, Martínez-López J. A Systems Biology- and Machine Learning-Based Study to Unravel Potential Therapeutic Mechanisms of Midostaurin as a Multitarget Therapy on FLT3-Mutated AML. BIOMEDINFORMATICS 2022; 2:375-397. [DOI: 10.3390/biomedinformatics2030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Acute myeloid leukemia (AML), a hematologic malignancy that results in bone marrow failure, is the most common acute leukemia in adults. The presence of FMS-related tyrosine kinase 3 (FLT3) mutations is associated with a poor prognosis, making the evaluation of FLT3-inhibitors an imperative goal in clinical trials. Midostaurin was the first FLT3-inhibitor approved by the FDA and EMA for the treatment of FLT3-mutated AML, and it showed a significant improvement in overall survival for newly diagnosed patients treated with midostaurin, in combination with standard chemotherapy (RATIFY study). The main interest of midostaurin has been the FLT3-specific inhibition, but little is known about its role as a multikinase inhibitor and whether it may be used in relapse and maintenance therapy. Here, we used systems biology- and machine learning-based approaches to deepen the potential benefits of the multitarget activity of midostaurin and to better understand its anti-leukemic effect on FLT3-mutated AML. The resulting in silico study revealed that the multikinase activity of midostaurin may play a role in the treatment’s efficacy. Additionally, we propose a series of molecular mechanisms that support a potential benefit of midostaurin as a maintenance therapy in FLT3-mutated AML, by regulating the microenvironment. The obtained results are backed up using independent gene expression data.
Collapse
Affiliation(s)
- Marina Díaz-Beyá
- Department of Hematology, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Myeloid Neoplasms Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - María García-Fortes
- Hematology Department, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Raquel Valls
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | - Laura Artigas
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | - Mª Teresa Gómez-Casares
- Hematology Service, Hospital Universitario Insular Materno-Infantil, 35016 Las Palmas de Gran Canaria, Spain
| | - Pau Montesinos
- Departament of Hematology, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Fermín Sánchez-Guijo
- Cancer Research Center (CIC), Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Mireia Coma
- Molecular Health Department, Anaxomics Biotech SL, 08007 Barcelona, Spain
| | | | - Joaquín Martínez-López
- Hospital 12 de Octubre. Universidad Complutense. CNIO. Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Instituto Carlos III, 28041 Madrid, Spain
| |
Collapse
|
47
|
Uslu M, Albayrak E, Kocabaş F. Competitive inhibition of IL-2/IL-2R has a dual effect on HSC ex vivo expansion and IL-2R (CD25) content. Int Immunopharmacol 2022; 110:109035. [PMID: 35834953 DOI: 10.1016/j.intimp.2022.109035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/27/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022]
Abstract
Interleukin-2 (IL-2) and its receptor play a pivotal role in the regulation of immune response and possess both immune-regulatory and immune-stimulatory functions. As a cytokine of lymphoid cells, the role of IL-2 has been revealed in hematopoietic stem cell (HSC) maintenance and proper hematopoiesis. Here, we investigated that small molecule Ro 26-4550 trifluoroacetate (Ro) mediated competitive inhibition of IL-2 and its receptor alpha subunit (IL-2Rα) throughout ex vivo culture. Ro treatment induced murine and human ex vivo expansion of hematopoietic stem and progenitor cells (HSPCs). Ro treated HSPCs sustained self-renewal ability and low apoptotic activity. As a competitive inhibitor of IL-2/IL-2Rα interaction, Ro small molecule induced human HSPCs to entry into cell cycle. The proliferation of bone marrow mesenchymal stem cells (MSC) and fibroblasts were also highly increased post treatment. Besides, Ro treatment enhanced IL-2Rα (CD25) expression independent of IL-2 administration in human mPB-derived HSPCs and BM-derived HSPCs. Increased IL-2Rα (CD25) expression in BM-HSPCs was associated with the increase in the CD4+CD25+ T cell population. Xenotransplantation of immunodeficient mice with ex vivo expanded human CD34+ cells after Ro treatment revealed an efficient multi-lineage reconstitution in the recipient. These findings shed light on the role of IL-2/IL-2Rα interaction in HSC expansion, in vivo and in vitro HSC self-renewal ability and repopulation capacity as well as a possible mean for the induction of CD25 expressing cells in hematopoietic compartments.
Collapse
Affiliation(s)
- Merve Uslu
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye; Johns Hopkins All Children's Hospital, USA
| | - Esra Albayrak
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye; Center of Stem Cell Research and Application, Ondokuz Mayıs University, Samsun, Türkiye
| | - Fatih Kocabaş
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye.
| |
Collapse
|
48
|
Niu J, Peng D, Liu L. Drug Resistance Mechanisms of Acute Myeloid Leukemia Stem Cells. Front Oncol 2022; 12:896426. [PMID: 35865470 PMCID: PMC9294245 DOI: 10.3389/fonc.2022.896426] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/06/2022] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukemia (AML) is a polyclonal and heterogeneous hematological malignancy. Relapse and refractory after induction chemotherapy are still challenges for curing AML. Leukemia stem cells (LSCs), accepted to originate from hematopoietic stem/precursor cells, are the main root of leukemogenesis and drug resistance. LSCs are dynamic derivations and possess various elusive resistance mechanisms. In this review, we summarized different primary resistance and remolding mechanisms of LSCs after chemotherapy, as well as the indispensable role of the bone marrow microenvironment on LSCs resistance. Through a detailed and comprehensive review of the spectacle of LSCs resistance, it can provide better strategies for future researches on eradicating LSCs and clinical treatment of AML.
Collapse
Affiliation(s)
| | | | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
49
|
He H, Wang Z, Yu H, Zhang G, Wen Y, Cai Z. Prioritizing risk genes as novel stratification biomarkers for acute monocytic leukemia by integrative analysis. Discov Oncol 2022; 13:55. [PMID: 35771283 PMCID: PMC9247126 DOI: 10.1007/s12672-022-00516-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/08/2022] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a blood cancer with high heterogeneity and stratified as M0-M7 subtypes in the French-American-British (FAB) diagnosis system. Improved diagnosis with leverage of key molecular inputs will assist precisive medicine. Through deep-analyzing the transcriptomic data and mutations of AML, we report that a modern clustering algorithm, t-distributed Stochastic Neighbor Embedding (t-SNE), successfully demarcates M2, M3 and M5 territories while M4 bias to M5 and M0 & M1 bias to M2, consistent with the traditional FAB classification. Combining with mutation profiles, the results show that top recurrent AML mutations were unbiasedly allocated into M2 and M5 territories, indicating the t-SNE instructed transcriptomic stratification profoundly outperforms mutation profiling in the FAB system. Further functional data mining prioritizes several myeloid-specific genes as potential regulators of AML progression and treatment by Venetoclax, a BCL2 inhibitor. Among them two encode membrane proteins, LILRB4 and LRRC25, which could be utilized as cell surface biomarkers for monocytic AML or for innovative immuno-therapy candidates in future. In summary, our deep functional data-mining analysis warrants several unappreciated immune signaling-encoding genes as novel diagnostic biomarkers and potential therapeutic targets.
Collapse
Affiliation(s)
- Hang He
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Zhiqin Wang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Hanzhi Yu
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Guorong Zhang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Yuchen Wen
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Zhigang Cai
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China.
- Department of Rheumatology, Tianjin Medical University General Hospital, Tianjin, China.
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
50
|
Dal Bello R, Pasanisi J, Joudinaud R, Duchmann M, Pardieu B, Ayaka P, Di Feo G, Sodaro G, Chauvel C, Kim R, Vasseur L, Chat L, Ling F, Pacchiardi K, Vaganay C, Berrou J, Benaksas C, Boissel N, Braun T, Preudhomme C, Dombret H, Raffoux E, Fenouille N, Clappier E, Adès L, Puissant A, Itzykson R. A multiparametric niche-like drug screening platform in acute myeloid leukemia. Blood Cancer J 2022; 12:95. [PMID: 35750691 PMCID: PMC9232632 DOI: 10.1038/s41408-022-00689-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/13/2022] [Accepted: 06/07/2022] [Indexed: 02/06/2023] Open
Abstract
Functional precision medicine in AML often relies on short-term in vitro drug sensitivity screening (DSS) of primary patient cells in standard culture conditions. We designed a niche-like DSS assay combining physiologic hypoxia (O2 3%) and mesenchymal stromal cell (MSC) co-culture with multiparameter flow cytometry to enumerate lymphocytes and differentiating (CD11/CD14/CD15+) or leukemic stem cell (LSC)-enriched (GPR56+) cells within the leukemic bulk. After functional validation of GPR56 expression as a surrogate for LSC enrichment, the assay identified three patterns of response, including cytotoxicity on blasts sparing LSCs, induction of differentiation, and selective impairment of LSCs. We refined our niche-like culture by including plasma-like amino-acid and cytokine concentrations identified by targeted metabolomics and proteomics of primary AML bone marrow plasma samples. Systematic interrogation revealed distinct contributions of each niche-like component to leukemic outgrowth and drug response. Short-term niche-like culture preserved clonal architecture and transcriptional states of primary leukemic cells. In a cohort of 45 AML samples enriched for NPM1c AML, the niche-like multiparametric assay could predict morphologically (p = 0.02) and molecular (NPM1c MRD, p = 0.04) response to anthracycline-cytarabine induction chemotherapy. In this cohort, a 23-drug screen nominated ruxolitinib as a sensitizer to anthracycline-cytarabine. This finding was validated in an NPM1c PDX model.
Collapse
Affiliation(s)
- Reinaldo Dal Bello
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Justine Pasanisi
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Romane Joudinaud
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Matthieu Duchmann
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Bryann Pardieu
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Paolo Ayaka
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Giuseppe Di Feo
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Gaetano Sodaro
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Clémentine Chauvel
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Rathana Kim
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Loic Vasseur
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Laureen Chat
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Frank Ling
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Kim Pacchiardi
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Camille Vaganay
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Jeannig Berrou
- Université Paris Cité, EA 3518, IRSL, Hôpital Saint-Louis, F-75010, Paris, France
| | - Chaima Benaksas
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Nicolas Boissel
- Service Hématologie Adolescents Jeunes Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Thorsten Braun
- Université Paris Cité, EA 3518, IRSL, Hôpital Saint-Louis, F-75010, Paris, France.,Service d'Hématologie clinique, Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, Bobigny, France
| | - Claude Preudhomme
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000, Lille, France
| | - Hervé Dombret
- Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France.,Université Paris Cité, EA 3518, IRSL, Hôpital Saint-Louis, F-75010, Paris, France
| | - Emmanuel Raffoux
- Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Nina Fenouille
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Emmanuelle Clappier
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.,Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Lionel Adès
- Service Hématologie Seniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Alexandre Puissant
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Raphael Itzykson
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France. .,Service Hématologie Adultes, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France.
| |
Collapse
|