1
|
Li J, Wang Y, Dong C, Luo L. Advancements in leukemia management: Bridging diagnosis, prognosis and nanotechnology (Review). Int J Oncol 2024; 65:112. [PMID: 39364739 DOI: 10.3892/ijo.2024.5700] [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: 04/19/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
Leukemia is a cancer that starts in blood stem cells in the bone marrow. Today, the proper diagnosis and prognosis of leukemia are essential in mitigating the morbidity and mortality associated with this malignancy. The advent of novel biomarkers, particularly those related to minimal residual disease, has paved the way for personalized therapeutic strategies and enables the quantitative assessment of patient responses to treatment regimens. Novel diagnostic and targeted drug delivery may be helpful for the improved management of leukemia. Genetic clinical parameters, such as chromosomal abnormalities, are crucial in diagnosing and guiding treatment decisions. These genetic markers also provide valuable prognostic information, helping to predict patient outcomes and tailor personalized treatment plans. In the present review, the studies on the diagnostic and prognostic parameters of leukemia were analyzed. The prognosis of leukemia was investigated in most of the studies, and the remaining were performed on diagnosis. The clinical and laboratory prognostic parameters were the most common, followed by diagnostic hematological parameters, diagnostic blood parameter studies, and diagnostic immunological parameters. Clinical and laboratory prognostic and hematologic parameters were the most extensively studied. The methods used to diagnose and prognose the leukemia cases in these studies were predominantly clinical hematology. Numerous surface proteins and receptors, including CD45, CD27, CD29, CD38, CD27, CD123, CD56 and CD25, react similarly in various kinds of leukemia, which are ideal for targeted drug delivery. Drug delivery to leukemia cells encounters several significant obstacles, including heterogeneity, that hinder the effectiveness of treatment. Nanocarriers play a critical role in targeted drug delivery for leukemia by enhancing the precision of treatments directed at surface proteins and receptors. Additionally, they can be functionalized with targeting drugs and antibodies to target specific tissues and cells.
Collapse
Affiliation(s)
- Jingbo Li
- Department of Ultrasound Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yingxue Wang
- Department of Ultrasound Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Chunli Dong
- Department of Critical Care Medicine, Jilin People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Lifu Luo
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
2
|
Prajapati SK, Kumari N, Bhowmik D, Gupta R. Recent advancements in biomarkers, therapeutics, and associated challenges in acute myeloid leukemia. Ann Hematol 2024; 103:4375-4400. [PMID: 39198271 DOI: 10.1007/s00277-024-05963-x] [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: 06/22/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Acute myeloid leukemia (AML) is a common type of leukemia that has a high mortality rate. The reasons for high mortality in patients with AML are therapeutic resistance, limited ability to predict duration of response, and likelihood of cancer relapse. Biomarkers, such as leukemic stem cell biomarkers, circulatory biomarkers, measurable residual disease biomarkers, and molecular biomarkers, are used for prognosis, diagnosis, and targeted killing to selectively eliminate AML cells. They also play an indispensable role in providing therapeutic resistance to patients with AML. Therefore, targeting these biomarkers will improve the outcome of AML patients. However, identifying biomarkers that can differentiate between treatment-responsive and non-responsive AML patients remains a challenge. This review discusses recent advancements in AML biomarkers, promising therapeutics, and associated challenges in the treatment of AML.
Collapse
Affiliation(s)
- Suresh Kumar Prajapati
- Research and Development Cell, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, India
| | - Neha Kumari
- Parul Institute of Applied Sciences, Parul University, Vadodara, 380060, India
| | - Doulat Bhowmik
- Parul Institute of Applied Sciences, Parul University, Vadodara, 380060, India
| | - Reeshu Gupta
- Research and Development Cell, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, India.
- Parul Institute of Applied Sciences, Parul University, Vadodara, 380060, India.
| |
Collapse
|
3
|
Arner A, Ettinger A, Blaser BW, Schmid B, Jeremias I, Rostam N, Binder-Blaser V. In vivo monitoring of leukemia-niche interactions in a zebrafish xenograft model. PLoS One 2024; 19:e0309415. [PMID: 39213296 PMCID: PMC11364250 DOI: 10.1371/journal.pone.0309415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common type of malignancy in children. ALL prognosis after initial diagnosis is generally good; however, patients suffering from relapse have a poor outcome. The tumor microenvironment is recognized as an important contributor to relapse, yet the cell-cell interactions involved are complex and difficult to study in traditional experimental models. In the present study, we established an innovative larval zebrafish xenotransplantation model, that allows the analysis of leukemic cells (LCs) within an orthotopic niche using time-lapse microscopic and flow cytometric approaches. LCs homed, engrafted and proliferated within the hematopoietic niche at the time of transplant, the caudal hematopoietic tissue (CHT). A specific dissemination pattern of LCs within the CHT was recorded, as they extravasated over time and formed clusters close to the dorsal aorta. Interactions of LCs with macrophages and endothelial cells could be quantitatively characterized. This zebrafish model will allow the quantitative analysis of LCs in a functional and complex microenvironment, to study mechanisms of niche mediated leukemogenesis, leukemia maintenance and relapse development.
Collapse
Affiliation(s)
- Anja Arner
- Department of Pediatric Hematology/Oncology, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University (LMU), Munich, Germany
| | - Andreas Ettinger
- Institute of Epigenetics and Stem Cells, Helmholtz Centre Munich, German Research Center for Environmental Health, Munich, Germany
| | - Bradley Wayne Blaser
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Bettina Schmid
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Irmela Jeremias
- Department of Pediatric Hematology/Oncology, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University (LMU), Munich, Germany
- Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Centre Munich, German Research Center for Environmental Health, Munich, Germany
| | - Nadia Rostam
- Department of Pediatric Hematology/Oncology, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University (LMU), Munich, Germany
- Department of Biology, University of Sulaimani, Sulaymaniyah, Iraq
| | - Vera Binder-Blaser
- Department of Pediatric Hematology/Oncology, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University (LMU), Munich, Germany
| |
Collapse
|
4
|
Osiriphan M, Insukhin C, Anuchapreeda S, Khamphikham P, Duangmano S. MicroRNA‑223 overexpression suppresses protein kinase C ε expression in human leukemia stem cell‑like KG‑1a cells. Mol Clin Oncol 2024; 21:48. [PMID: 38881704 PMCID: PMC11176719 DOI: 10.3892/mco.2024.2746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/01/2024] [Indexed: 06/18/2024] Open
Abstract
MicroRNA-223 (miR-223) is dysregulated in various cancer types, including acute myeloid leukemia (AML). Despite this, there has been a lack of studies exploring the role of miR-223 in leukemic stem cells, particularly those involved in drug resistance, a major cause of chemotherapy failure in AML. The present study aimed to elucidate the impact of miR-223 on drug resistance in the leukemic stem-cell line, KG-1a. Two AML cell lines, KG-1 and KG-1a, differing in the proportion of CD34+CD38- cells, were assessed for doxorubicin (DOX) sensitivity using the Cell Counting Kit-8 assay. The expression levels of miR-223 and protein kinase C ε (PKCε) were evaluated via reverse transcription-quantitative PCR and western blot analysis. The association between miR-223 and its target, PKCε, was confirmed by luciferase activity assay. The effects of miR-223 overexpression and PKCε inhibition were also evaluated in KG-1a cells using miR-223 mimic and small interfering (si)RNA transfection, respectively. Daunorubicin was then used to assess drug sensitivity in the siRNA-transfected KG-1a cells. Compared with KG-1 cells, KG-1a cells displayed greater resistance to DOX, and had increased PKCε levels and decreased miR-223 expression. Overexpression of miR-223 led to PKCε protein downregulation in KG-1a cells, which was further confirmed by a luciferase assay demonstrating miR-223 targeting of PKCε. However, despite these effects, miR-223 overexpression and PKCε inhibition did not change drug sensitivity in KG-1a cells compared with negative control cells. In summary, the present study demonstrated that miR-223 could target and silence PKCε expression in KG-1a cells; however, the chemoresistance of KG-1a cells to anthracycline drugs may not be directly associated with the low expression of miR-223.
Collapse
Affiliation(s)
- Mallika Osiriphan
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
- Master's Degree Program in Medical Technology (under the Chiang Mai University Presidential Scholarship), Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
| | - Charapat Insukhin
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
| | - Songyot Anuchapreeda
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
- Cancer Research Unit of Associated Medical Sciences, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
- Hematology and Health Technology Research Center, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
| | - Pinyaphat Khamphikham
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
- Hematology and Health Technology Research Center, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
| | - Suwit Duangmano
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
- Cancer Research Unit of Associated Medical Sciences, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
- Hematology and Health Technology Research Center, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Mueang Chiang Mai, Chiang Mai 50200, Thailand
| |
Collapse
|
5
|
Vicente ATS, Salvador JAR. PROteolysis-Targeting Chimeras (PROTACs) in leukemia: overview and future perspectives. MedComm (Beijing) 2024; 5:e575. [PMID: 38845697 PMCID: PMC11154823 DOI: 10.1002/mco2.575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
Leukemia is a heterogeneous group of life-threatening malignant disorders of the hematopoietic system. Immunotherapy, radiotherapy, stem cell transplantation, targeted therapy, and chemotherapy are among the approved leukemia treatments. Unfortunately, therapeutic resistance, side effects, relapses, and long-term sequelae occur in a significant proportion of patients and severely compromise the treatment efficacy. The development of novel approaches to improve outcomes is therefore an unmet need. Recently, novel leukemia drug discovery strategies, including targeted protein degradation, have shown potential to advance the field of personalized medicine for leukemia patients. Specifically, PROteolysis-TArgeting Chimeras (PROTACs) are revolutionary compounds that allow the selective degradation of a protein by the ubiquitin-proteasome system. Developed against a wide range of cancer targets, they show promising potential in overcoming many of the drawbacks associated with conventional therapies. Following the exponential growth of antileukemic PROTACs, this article reviews PROTAC-mediated degradation of leukemia-associated targets. Chemical structures, in vitro and in vivo activities, pharmacokinetics, pharmacodynamics, and clinical trials of PROTACs are critically discussed. Furthermore, advantages, challenges, and future perspectives of PROTACs in leukemia are covered, in order to understand the potential that these novel compounds may have as future drugs for leukemia treatment.
Collapse
Affiliation(s)
- André T. S. Vicente
- Laboratory of Pharmaceutical ChemistryFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
- Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical ChemistryFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
- Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
| |
Collapse
|
6
|
Capelli D. FLT3-Mutated Leukemic Stem Cells: Mechanisms of Resistance and New Therapeutic Targets. Cancers (Basel) 2024; 16:1819. [PMID: 38791898 PMCID: PMC11119130 DOI: 10.3390/cancers16101819] [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: 03/18/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Despite the availability of target drugs in the first and second line, only 30% of FLT3mut AMLs are cured. Among the multiple mechanisms of resistance, those of FLT3mut LSC are the most difficult to eradicate because of their metabolic and genomic characteristics. Reactivation of glycogen synthesis, inhibition of the RAS/MAPK pathway, and degradation of FLT3 may be potential aids to fight the resistance of LSC to FLT3i. LSC is also characterized by the expression of a CD34+/CD25+/CD123+/CD99+ immunophenotype. The receptor and ligand of FLT3, the natural killer group 2 member D ligand (NKGD2L), and CD123 are some of the targets of chimeric antigen receptor T cells (CAR-T), bispecific T-cell engager molecules (BiTEs), CAR-NK and nanoparticles recently designed and reported here. The combination of these new therapeutic options, hopefully in a minimal residual disease (MRD)-driven approach, could provide the future answer to the challenge of treating FLT3mut AML.
Collapse
Affiliation(s)
- Debora Capelli
- Department of Hematology, Azienda Ospedaliera Universitaria, Ospedali Riuniti di Ancona, Via Conca 71, 60126 Ancona, Italy
| |
Collapse
|
7
|
Murtadha M, Park M, Zhu Y, Caserta E, Napolitano O, Tandoh T, Moloudizargari M, Pozhitkov A, Singer M, Dona AA, Vahed H, Gonzalez A, Ly K, Ouyang C, Sanchez JF, Nigam L, Duplan A, Chowdhury A, Ghoda L, Li L, Zhang B, Krishnan A, Marcucci G, Williams JC, Pichiorri F. A CD38-directed, single-chain T-cell engager targets leukemia stem cells through IFN-γ-induced CD38 expression. Blood 2024; 143:1599-1615. [PMID: 38394668 PMCID: PMC11103097 DOI: 10.1182/blood.2023021570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 02/25/2024] Open
Abstract
ABSTRACT Treatment resistance of leukemia stem cells (LSCs) and suppression of the autologous immune system represent major challenges to achieve a cure in acute myeloid leukemia (AML). Although AML blasts generally retain high levels of surface CD38 (CD38pos), LSCs are frequently enriched in the CD34posCD38neg blast fraction. Here, we report that interferon gamma (IFN-γ) reduces LSCs clonogenic activity and induces CD38 upregulation in both CD38pos and CD38neg LSC-enriched blasts. IFN-γ-induced CD38 upregulation depends on interferon regulatory factor 1 transcriptional activation of the CD38 promoter. To leverage this observation, we created a novel compact, single-chain CD38-CD3 T-cell engager (BN-CD38) designed to promote an effective immunological synapse between CD38pos AML cells and both CD8pos and CD4pos T cells. We demonstrate that BN-CD38 engages autologous CD4pos and CD8pos T cells and CD38pos AML blasts, leading to T-cell activation and expansion and to the elimination of leukemia cells in an autologous setting. Importantly, BN-CD38 engagement induces the release of high levels of IFN-γ, driving the expression of CD38 on CD34posCD38neg LSC-enriched blasts and their subsequent elimination. Critically, although BN-CD38 showed significant in vivo efficacy across multiple disseminated AML cell lines and patient-derived xenograft models, it did not affect normal hematopoietic stem cell clonogenicity and the development of multilineage human immune cells in CD34pos humanized mice. Taken together, this study provides important insights to target and eliminate AML LSCs.
Collapse
Affiliation(s)
- Mariam Murtadha
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Miso Park
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Yinghui Zhu
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
- Research Center for Translational Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Enrico Caserta
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ottavio Napolitano
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Theophilus Tandoh
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Milad Moloudizargari
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Alex Pozhitkov
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Mahmoud Singer
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ada Alice Dona
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Hawa Vahed
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Asaul Gonzalez
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Kevin Ly
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ching Ouyang
- Integrative Genomics Core, City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA
| | - James F. Sanchez
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
| | - Lokesh Nigam
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Amanda Duplan
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Arnab Chowdhury
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Lucy Ghoda
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ling Li
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Bin Zhang
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
| | - Guido Marcucci
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - John C. Williams
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Flavia Pichiorri
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| |
Collapse
|
8
|
Ruszkowska-Ciastek B, Kwiatkowska K, Marques-da-Silva D, Lagoa R. Cancer Stem Cells from Definition to Detection and Targeted Drugs. Int J Mol Sci 2024; 25:3903. [PMID: 38612718 PMCID: PMC11011379 DOI: 10.3390/ijms25073903] [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: 02/26/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Cancers remain the second leading cause of mortality in the world. Preclinical and clinical studies point an important role of cancer/leukaemia stem cells (CSCs/LSCs) in the colonisation at secondary organ sites upon metastatic spreading, although the precise mechanisms for specific actions are still not fully understood. Reviewing the present knowledge on the crucial role of CSCs/LSCs, their plasticity, and population heterogeneity in treatment failures in cancer patients is timely. Standard chemotherapy, which acts mainly on rapidly dividing cells, is unable to adequately affect CSCs with a low proliferation rate. One of the proposed mechanisms of CSC resistance to anticancer agents is the fact that these cells can easily shift between different phases of the cell cycle in response to typical cell stimuli induced by anticancer drugs. In this work, we reviewed the recent studies on CSC/LSC alterations associated with disease recurrence, and we systematised the functional assays, markers, and novel methods for CSCs screening. This review emphasises CSCs' involvement in cancer progression and metastasis, as well as CSC/LSC targeting by synthetic and natural compounds aiming at their elimination or modulation of stemness properties.
Collapse
Affiliation(s)
- Barbara Ruszkowska-Ciastek
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University, Collegium Medicum, 85-094 Bydgoszcz, Poland
| | - Katarzyna Kwiatkowska
- Department of Laboratory Diagnostics, Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland;
| | - Dorinda Marques-da-Silva
- Laboratory of Separation and Reaction Engineering-Laboratory of Catalysis and Materials (LSRE-LCM), Polytechnic Institute of Leiria, 2411-901 Leiria, Portugal; (D.M.-d.-S.); (R.L.)
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Ricardo Lagoa
- Laboratory of Separation and Reaction Engineering-Laboratory of Catalysis and Materials (LSRE-LCM), Polytechnic Institute of Leiria, 2411-901 Leiria, Portugal; (D.M.-d.-S.); (R.L.)
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| |
Collapse
|
9
|
Azizidoost S, Nasrolahi A, Sheykhi-Sabzehpoush M, Anbiyaiee A, Khoshnam SE, Farzaneh M, Uddin S. Signaling pathways governing the behaviors of leukemia stem cells. Genes Dis 2024; 11:830-846. [PMID: 37692500 PMCID: PMC10491880 DOI: 10.1016/j.gendis.2023.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 01/02/2023] [Indexed: 08/28/2023] Open
Abstract
Leukemia is a malignancy in the blood that develops from the lymphatic system and bone marrow. Although various treatment options have been used for different types of leukemia, understanding the molecular pathways involved in the development and progression of leukemia is necessary. Recent studies showed that leukemia stem cells (LSCs) play essential roles in the pathogenesis of leukemia by targeting several signaling pathways, including Notch, Wnt, Hedgehog, and STAT3. LSCs are highly proliferative cells that stimulate tumor initiation, migration, EMT, and drug resistance. This review summarizes cellular pathways that stimulate and prevent LSCs' self-renewal, metastasis, and tumorigenesis.
Collapse
Affiliation(s)
- Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Mohadeseh Sheykhi-Sabzehpoush
- Department of Laboratory, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran 2193672411, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6193673111, Iran
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
10
|
Khorashad JS, Rizzo S, Tonks A. Reactive oxygen species and its role in pathogenesis and resistance to therapy in acute myeloid leukemia. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:5. [PMID: 38434766 PMCID: PMC10905166 DOI: 10.20517/cdr.2023.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/24/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Relapse following a short clinical response to therapy is the major challenge for the management of acute myeloid leukemia (AML) patients. Leukemic stem cells (LSC), as the source of relapse, have been investigated for their metabolic preferences and their alterations at the time of relapse. As LSC rely on oxidative phosphorylation (OXPHOS) for energy requirement, reactive oxygen species (ROS), as by-products of OXPHOS, have been investigated for their role in the effectiveness of the standard AML therapy. Increased levels of non-mitochondrial ROS, generated by nicotinamide adenine dinucleotide phosphate oxidase, in a subgroup of AML patients add to the complexity of studying ROS. Although there are various studies presenting the contribution of ROS to AML pathogenesis, resistance, and its inhibition or activation as a target, a model that can clearly explain its role in AML has not been conceptualized. This is due to the heterogeneity of AML, the dynamics of ROS production, which is influenced by factors such as the type of treatment, cell differentiation state, mitochondrial activity, and also the heterogeneous generation of non-mitochondrial ROS and limited available data on their interaction with the microenvironment. This review summarizes these challenges and the recent progress in this field.
Collapse
Affiliation(s)
- Jamshid Sorouri Khorashad
- Department of Immunology and inflammation, Imperial College London, London, W12 0NN, UK
- Department of Molecular Pathology, Institute of Cancer Research, Sutton, SM2 5PT, UK
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sian Rizzo
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Alex Tonks
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
11
|
Tarui N. [The quick screening of binding compounds and proteins for drug discovery and pharmacological research]. Nihon Yakurigaku Zasshi 2024; 159:407-412. [PMID: 39496418 DOI: 10.1254/fpj.24057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
In drug discovery and pharmacological research, early identification of target molecules for compounds with pharmacological effects is crucial. However, this process often requires significant effort and can be rate-limiting, thereby slowing down research progress. This paper introduces a simplified and rapid method for quick screening of binding compounds or proteins. Utilizing Affinity Selection Mass Spectrometry (ASMS), this technique efficiently detects compound-target binding through size-exclusion chromatography and mass spectrometry. ASMS offers high sensitivity and specificity, making it ideal for accurate identification of binding interactions. We have further enhanced ASMS to handle membrane proteins without solubilization, creating Binder Selection Technology (BST). BST allows screening against both soluble and challenging membrane proteins such as GPCRs and SLC transporters. By using cell membrane fractions or organelle fractions with high target molecule expression, BST efficiently identifies potential binding compounds. This innovative method constructs a comprehensive database of binding compounds for various targets, facilitating rapid hypothesis testing and pharmacological evaluation. Additionally, BST screens 17,000 proteins, including membrane proteins, using wheat germ cell-free and animal cell expression systems. This approach allows exploration of binding interactions without labeling compounds or immobilizing proteins, preserving their native state. BST is powerful for identifying targets of compounds with known pharmacological effects but unknown targets in animal or cell-based assays. By utilizing BST, researchers can overcome bottlenecks in early drug discovery, significantly enhancing research speed and success rates. This method represents a major advancement, providing an efficient and effective way to identify and validate target molecules in drug discovery.
Collapse
|
12
|
Rezaee A, Ahmadpour S, Jafari A, Aghili S, Zadeh SST, Rajabi A, Raisi A, Hamblin MR, Mahjoubin-Tehran M, Derakhshan M. MicroRNAs, long non-coding RNAs, and circular RNAs and gynecological cancers: focus on metastasis. Front Oncol 2023; 13:1215194. [PMID: 37854681 PMCID: PMC10580988 DOI: 10.3389/fonc.2023.1215194] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/28/2023] [Indexed: 10/20/2023] Open
Abstract
Gynecologic cancer is a significant cause of death in women worldwide, with cervical cancer, ovarian cancer, and endometrial cancer being among the most well-known types. The initiation and progression of gynecologic cancers involve a variety of biological functions, including angiogenesis and metastasis-given that death mostly occurs from metastatic tumors that have invaded the surrounding tissues. Therefore, understanding the molecular pathways underlying gynecologic cancer metastasis is critical for enhancing patient survival and outcomes. Recent research has revealed the contribution of numerous non-coding RNAs (ncRNAs) to metastasis and invasion of gynecologic cancer by affecting specific cellular pathways. This review focuses on three types of gynecologic cancer (ovarian, endometrial, and cervical) and three kinds of ncRNAs (long non-coding RNAs, microRNAs, and circular RNAs). We summarize the detailed role of non-coding RNAs in the different pathways and molecular interactions involved in the invasion and metastasis of these cancers.
Collapse
Affiliation(s)
- Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Ahmadpour
- Biotechnology Department, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Ameneh Jafari
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sarehnaz Aghili
- Department of Gynecology and Obstetrics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Maryam Mahjoubin-Tehran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Derakhshan
- Shahid Beheshti Fertility Clinic, Department of Gynecology and Obsteterics, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
13
|
Zhang Y, Jiang S, He F, Tian Y, Hu H, Gao L, Zhang L, Chen A, Hu Y, Fan L, Yang C, Zhou B, Liu D, Zhou Z, Su Y, Qin L, Wang Y, He H, Lu J, Xiao P, Hu S, Wang QF. Single-cell transcriptomics reveals multiple chemoresistant properties in leukemic stem and progenitor cells in pediatric AML. Genome Biol 2023; 24:199. [PMID: 37653425 PMCID: PMC10472599 DOI: 10.1186/s13059-023-03031-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/02/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Cancer patients can achieve dramatic responses to chemotherapy yet retain resistant tumor cells, which ultimately results in relapse. Although xenograft model studies have identified several cellular and molecular features that are associated with chemoresistance in acute myeloid leukemia (AML), to what extent AML patients exhibit these properties remains largely unknown. RESULTS We apply single-cell RNA sequencing to paired pre- and post-chemotherapy whole bone marrow samples obtained from 13 pediatric AML patients who had achieved disease remission, and distinguish AML clusters from normal cells based on their unique transcriptomic profiles. Approximately 50% of leukemic stem and progenitor populations actively express leukemia stem cell (LSC) and oxidative phosphorylation (OXPHOS) signatures, respectively. These clusters have a higher chance of tolerating therapy and exhibit an enhanced metabolic program in response to treatment. Interestingly, the transmembrane receptor CD69 is highly expressed in chemoresistant hematopoietic stem cell (HSC)-like populations (named the CD69+ HSC-like subpopulation). Furthermore, overexpression of CD69 results in suppression of the mTOR signaling pathway and promotion of cell quiescence and adhesion in vitro. Finally, the presence of CD69+ HSC-like cells is associated with unfavorable genetic mutations, the persistence of residual tumor cells in chemotherapy, and poor outcomes in independent pediatric and adult public AML cohorts. CONCLUSIONS Our analysis reveals leukemia stem cell and OXPHOS as two major chemoresistant features in human AML patients. CD69 may serve as a potential biomarker in defining a subpopulation of chemoresistant leukemia stem cells. These findings have important implications for targeting residual chemo-surviving AML cells.
Collapse
Affiliation(s)
- Yongping Zhang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Shuting Jiang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fuhong He
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanyuan Tian
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Haiyang Hu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li Gao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Lin Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Aili Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yixin Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Liyan Fan
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Chun Yang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Bi Zhou
- SuZhou Hospital of Anhui Medical University, Suzhou, China
| | - Dan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Zihan Zhou
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanxun Su
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Qin
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Hailong He
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Jun Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Peifang Xiao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Shaoyan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Qian-Fei Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
14
|
Yue S, An J, Zhang Y, Li J, Zhao C, Liu J, Liang L, Sun H, Xu Y, Zhong Z. Exogenous Antigen Upregulation Empowers Antibody Targeted Nanochemotherapy of Leukemia. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209984. [PMID: 37321606 DOI: 10.1002/adma.202209984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 05/12/2023] [Indexed: 06/17/2023]
Abstract
Acute myeloid leukemia (AML) is afflicted by a high-mortality rate and few treatment options. The lack of specific surface antigens severely hampers the development of targeted therapeutics and cell therapy. Here, it is shown that exogenous all-trans retinoic acid (ATRA) mediates selective and transient CD38 upregulation on leukemia cells by up to 20-fold, which enables high-efficiency targeted nanochemotherapy of leukemia with daratumumab antibody-directed polymersomal vincristine sulfate (DPV). Strikingly, treatment of two CD38-low expressing AML orthotopic models with ATRA and DPV portfolio strategies effectively eliminates circulating leukemia cells and leukemia invasion into bone marrow and organs, leading to exceptional survival benefits with 20-40% of mice becoming leukemia-free. The combination of exogenous CD38 upregulation and antibody-directed nanotherapeutics provides a unique and powerful targeted therapy for leukemia.
Collapse
Affiliation(s)
- Shujing Yue
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Jingnan An
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Yifan Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Jiaying Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, 215007, P. R. China
| | - Cenzhu Zhao
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Jingyi Liu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Lanlan Liang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Huanli Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yang Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
15
|
Yue M, Guo T, Nie DY, Zhu YX, Lin M. Advances of nanotechnology applied to cancer stem cells. World J Stem Cells 2023; 15:514-529. [PMID: 37424953 PMCID: PMC10324502 DOI: 10.4252/wjsc.v15.i6.514] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/01/2023] [Accepted: 04/18/2023] [Indexed: 06/26/2023] Open
Abstract
Cancer stem cells (CSCs) are a small proportion of the cells that exist in cancer tissues. They are considered to be the culprit of tumor genesis, development, drug resistance, metastasis and recurrence because of their self-renewal, proliferation, and differentiation potential. The elimination of CSCs is thus the key to cure cancer, and targeting CSCs provides a new method for tumor treatment. Due to the advantages of controlled sustained release, targeting and high biocompatibility, a variety of nanomaterials are used in the diagnosis and treatments targeting CSCs and promote the recognition and removal of tumor cells and CSCs. This article mainly reviews the research progress of nanotechnology in sorting CSCs and nanodrug delivery systems targeting CSCs. Furthermore, we identify the problems and future research directions of nanotechnology in CSC therapy. We hope that this review will provide guidance for the design of nanotechnology as a drug carrier so that it can be used in clinic for cancer therapy as soon as possible.
Collapse
Affiliation(s)
- Miao Yue
- Clinical Laboratory, Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China
| | - Ting Guo
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Deng-Yun Nie
- Clinical Laboratory, Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China
| | - Yin-Xing Zhu
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Mei Lin
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| |
Collapse
|
16
|
Murtadha M, Park M, Zhu Y, Caserta E, Dona AA, Singer M, Vahed H, Tasndoh T, Gonzalez A, Ly K, Sanchez JF, Chowdhury A, Pozhitkov A, Ghoda L, Li L, Zhang B, Krishnan A, Marcucci G, Williams J, Pichiorri F. Leveraging IFNγ/CD38 regulation to unmask and target leukemia stem cells in acute myelogenous leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530273. [PMID: 36909542 PMCID: PMC10002674 DOI: 10.1101/2023.02.27.530273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Elimination of drug-resistant leukemia stem cells (LSCs) represents a major challenge to achieve a cure in acute myeloid leukemia (AML). Although AML blasts generally retain high levels of surface CD38 (CD38pos), the presence of CD34 and lack of CD38 expression (CD34posCD38neg) are immunophenotypic features of both LSC-enriched AML blasts and normal hematopoietic stem cells (HSCs). We report that IFN-γ induces CD38 upregulation in LSC-enriched CD34posCD38neg AML blasts, but not in CD34posCD38neg HSCs. To leverage the IFN-γ mediated CD38 up-regulation in LSCs for clinical application, we created a compact, single-chain CD38-CD3-T cell engager (CD38-BIONIC) able to direct T cells against CD38pos blasts. Activated CD4pos and CD8pos T cells not only kill AML blasts but also produce IFNγ, which leads to CD38 expression on CD34posCD38neg LSC-enriched blasts. These cells then become CD38-BIONIC targets. The net result is an immune-mediated killing of both CD38neg and CD38pos AML blasts, which culminates in LSC depletion.
Collapse
Affiliation(s)
- Mariam Murtadha
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Miso Park
- Department of Molecular Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Yinghui Zhu
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Enrico Caserta
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Ada Alice Dona
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Mahmoud Singer
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Hawa Vahed
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Theophilus Tasndoh
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Asaul Gonzalez
- Department of Molecular Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Kevin Ly
- Department of Molecular Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - James F Sanchez
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
| | - Arnab Chowdhury
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Alex Pozhitkov
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Lucy Ghoda
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Ling Li
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Bin Zhang
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Amrita Krishnan
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
| | - Guido Marcucci
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - John Williams
- Department of Molecular Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Flavia Pichiorri
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| |
Collapse
|
17
|
Doghish AS, Abulsoud AI, Elshaer SS, Abdelmaksoud NM, Zaki MB, El-Mahdy HA, Ismail A, Fathi D, Elsakka EGE. miRNAs as cornerstones in chronic lymphocytic leukemia pathogenesis and therapeutic resistance- An emphasis on the interaction of signaling pathways. Pathol Res Pract 2023; 243:154363. [PMID: 36764011 DOI: 10.1016/j.prp.2023.154363] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Chronic lymphocytic leukemia (CLL) accounts for the vast majority of cases of leukemia. Patients of advanced age are more likely to develop the condition, which has a highly varied clinical course. Consideration of illness features and preceding treatment sequence, as well as patient preferences and comorbidities, is necessary for selecting the appropriate treatment for the appropriate patient. Therefore, there is an urgent need for novel biomarkers with high sensitivity and specificity to detect CLL early, monitor CLL patients, select the treatment responders, and reduce ineffective treatment, unwanted side effects, and unnecessary expenses. In both homeostasis and illness, microRNAs (miRNAs/miRs) play a vital role as master regulators of gene expression and, by extension, protein expression. MiRNAs typically reduce the stability of mRNAs, including those encoding genes involved in tumorigenesis processes as cell cycle regulation, inflammation, stress response, angiogenesis, differentiation, apoptosis, and invasion. Due to their unique properties, miRNAs are rapidly being exploited as accurate biomarkers for illness detection, and medicines based on miRNA targets are finding widespread application in clinical practice. Accordingly, the current review serves as a quick primer on CLL and the biogenesis of miRNAs. In addition to providing a brief overview of the miRNAs whose function in the progression of CLL has been established by recent in vitro or in vivo research through articulating the influence of these miRNAs on a wide variety of cellular functions, including increased proliferative potential; support for angiogenesis; cell cycle aberration; evasion of apoptosis; promotion of metastasis; and reduced sensitivity to specific treatments.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt; Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Shereen Saeid Elshaer
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt; Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11823, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| | - Doaa Fathi
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| |
Collapse
|
18
|
Brahmbhatt J, Kumar SP, Bhadresha K, Patel M, Rawal R. Targeting leukemic stem cell subpopulation in AML using phytochemicals: An in-silico and in-vitro approach. Comput Biol Med 2023; 155:106644. [PMID: 36774886 DOI: 10.1016/j.compbiomed.2023.106644] [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: 08/22/2022] [Revised: 01/10/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
It has been indicated that leukemic stem cells (LSCs), a subset of leukaemia cells, are responsible for therapy resistance and relapse in acute myeloid leukaemia (AML). Therefore, the current study aimed to discover an LSC biomarker in AML patients and identify a natural compound that may target the same. By performing the different gene expression analyses, we identified 12 up-regulated and 192 down-regulated genes in LSCs of AML compared to normal bone marrow-derived HSCs. Further STRING interaction, GO enrichment and KEGG pathway analysis were carried out to top hub genes. Wilms' tumour-1 (WT1) transcription factor was pointed out as the top hub gene and a potential biomarker for LSCs in AML. For the targeted inhibition of WT1, we performed screening and stimulation of potential natural compounds. The results revealed Gallic acid (GA) and Chlorogenic acid (CA) as promising WT1 inhibitors. In-vitro validation of cytotoxic effects of both GA and CA on THP-1 and HL-60 cell lines suggested that both these compounds inhibited cell proliferation. Still, GA has a more cytotoxic effect compared to CA. Next, we performed cell cycle analysis and apoptosis analysis and found that both compounds arrested cells in G0/G1 phase and induced apoptosis in both cell lines. Surprisingly, a significant decrease in colony formation and cell migration was also observed. However, GA gave more promising results in all cellular assays than CA. Furthermore, we studied the mRNA expression of WT1 and BCL2, which are transcriptionally activated by it. We found that GA significantly downregulated both these genes compared to CA. Our results suggested that GA is a potential inhibitor of WT1 and might be an excellent anti-LSCs natural drug for AML patients.
Collapse
Affiliation(s)
- Jpan Brahmbhatt
- Department of Life Sciences, School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Sivakumar Prasanth Kumar
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Kinjal Bhadresha
- Department of Life Sciences, School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Maulikkumar Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Rakesh Rawal
- Department of Life Sciences, School of Sciences, Gujarat University, Ahmedabad, 380009, India.
| |
Collapse
|
19
|
Teixeira A, Carreira L, Abalde-Cela S, Sampaio-Marques B, Areias AC, Ludovico P, Diéguez L. Current and Emerging Techniques for Diagnosis and MRD Detection in AML: A Comprehensive Narrative Review. Cancers (Basel) 2023; 15:cancers15051362. [PMID: 36900154 PMCID: PMC10000116 DOI: 10.3390/cancers15051362] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Acute myeloid leukemia (AML) comprises a group of hematologic neoplasms characterized by abnormal differentiation and proliferation of myeloid progenitor cells. AML is associated with poor outcome due to the lack of efficient therapies and early diagnostic tools. The current gold standard diagnostic tools are based on bone marrow biopsy. These biopsies, apart from being very invasive, painful, and costly, have low sensitivity. Despite the progress uncovering the molecular pathogenesis of AML, the development of novel detection strategies is still poorly explored. This is particularly important for patients that check the criteria for complete remission after treatment, since they can relapse through the persistence of some leukemic stem cells. This condition, recently named as measurable residual disease (MRD), has severe consequences for disease progression. Hence, an early and accurate diagnosis of MRD would allow an appropriate therapy to be tailored, improving a patient's prognosis. Many novel techniques with high potential in disease prevention and early detection are being explored. Among them, microfluidics has flourished in recent years due to its ability at processing complex samples as well as its demonstrated capacity to isolate rare cells from biological fluids. In parallel, surface-enhanced Raman scattering (SERS) spectroscopy has shown outstanding sensitivity and capability for multiplex quantitative detection of disease biomarkers. Together, these technologies can allow early and cost-effective disease detection as well as contribute to monitoring the efficiency of treatments. In this review, we aim to provide a comprehensive overview of AML disease, the conventional techniques currently used for its diagnosis, classification (recently updated in September 2022), and treatment selection, and we also aim to present how novel technologies can be applied to improve the detection and monitoring of MRD.
Collapse
Affiliation(s)
- Alexandra Teixeira
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Luís Carreira
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
| | - Sara Abalde-Cela
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Anabela C. Areias
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (P.L.); (L.D.)
| | - Lorena Diéguez
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
- Correspondence: (P.L.); (L.D.)
| |
Collapse
|
20
|
Farzaneh M, Najafi S, Sheykhi-Sabzehpoush M, Nezhad Dehbashi F, Anbiyaee O, Nasrolahi A, Azizidoost S. The stem cell-specific long non-coding RNAs in leukemia. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:345-351. [PMID: 36168086 DOI: 10.1007/s12094-022-02952-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/09/2022] [Indexed: 01/27/2023]
Abstract
Leukemia is defined as a heterogeneous group of hematological cancers whose prevalence is on the rise worldwide. Despite the large body of studies, the etiology of leukemia has not been fully elucidated. Leukemia stem cells (LSCs) are a subpopulation of cancer cells that sustain the growth of the leukemic clone and are the main culprit for the maintenance of the neoplasm. In contrast to most leukemia cells, LSCs are resistant to chemo- and radiotherapy. Several recent studies demonstrated the altered expression profile of long non-coding RNAs (lncRNAs) in LSCs and shed light on the role of lncRNAs in the survival, proliferation, and differentiation of LSCs. LncRNAs are transcripts longer than 200 nucleotides that are implicated in several cellular and molecular processes such as gene expression, apoptosis, and carcinogenesis. Likewise, lncRNAs have shown a prognostic marker in leukemia patients and represent novel treatment options. Herein, we review the current knowledge concerning lncRNAs' implication in the pathogenesis of LSCs and discuss their prognostic, diagnostic, and therapeutic potential.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Fereshteh Nezhad Dehbashi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Omid Anbiyaee
- Cardiovascular Research Center, School of Medicine, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ava Nasrolahi
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
21
|
Cerneckis J, Cui Q, Liu W, Shi Y. RNA Modifications in Cancer Stem Cell Biology. Cancer Treat Res 2023; 190:25-47. [PMID: 38112998 DOI: 10.1007/978-3-031-45654-1_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] [Indexed: 12/21/2023]
Abstract
Post-transcriptional regulation of gene expression shapes the cell state both in health and disease. RNA modifications-especially N6-methyladenosine (m6A)-have recently emerged as key players in RNA processing that depends on a sophisticated interplay between proteins of the RNA modification machinery. Importantly, the RNA epitranscriptome becomes dysregulated in cancer and promotes cancer-associated gene expression programs as well as cancer cell adaptation to the tumor microenvironment. At the top of the tumor hierarchy, cancer stem cells (CSCs) are master regulators of tumorigenesis and resistance to therapeutic intervention. Therefore, defining how RNA modifications influence the CSC state is of great importance for cancer drug development. In this chapter, we summarize the current knowledge of the roles of RNA modifications in shaping the CSC state and driving gene expression programs that confer stem-like properties to CSCs, promote CSC adaptation to the local microenvironment, and endow CSCs with metastatic potential and drug resistance.
Collapse
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Qi Cui
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Wei Liu
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Department of Immunology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
22
|
Gu L, Liao P, Liu H. Cancer-associated fibroblasts in acute leukemia. Front Oncol 2022; 12:1022979. [PMID: 36601484 PMCID: PMC9806275 DOI: 10.3389/fonc.2022.1022979] [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: 08/19/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Although the prognosis for acute leukemia has greatly improved, treatment of relapsed/refractory acute leukemia (R/R AL) remains challenging. Recently, increasing evidence indicates that the bone marrow microenvironment (BMM) plays a crucial role in leukemogenesis and therapeutic resistance; therefore, BMM-targeted strategies should be a potent protocol for treating R/R AL. The targeting of cancer-associated fibroblasts (CAFs) in solid tumors has received much attention and has achieved some progress, as CAFs might act as an organizer in the tumor microenvironment. Additionally, over the last 10 years, attention has been drawn to the role of CAFs in the BMM. In spite of certain successes in preclinical and clinical studies, the heterogeneity and plasticity of CAFs mean targeting them is a big challenge. Herein, we review the heterogeneity and roles of CAFs in the BMM and highlight the challenges and opportunities associated with acute leukemia therapies that involve the targeting of CAFs.
Collapse
Affiliation(s)
- Ling Gu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China,The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China,*Correspondence: Ling Gu, ; Ping Liao, ; Hanmin Liu,
| | - Ping Liao
- Calcium Signalling Laboratory, National Neuroscience Institute, Singapore, Singapore,Academic & Clinical Development, Duke-NUS Medical School, Singapore, Singapore,Health and Social Sciences, Singapore Institute of Technology, Singapore, Singapore,*Correspondence: Ling Gu, ; Ping Liao, ; Hanmin Liu,
| | - Hanmin Liu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China,The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China,Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China,*Correspondence: Ling Gu, ; Ping Liao, ; Hanmin Liu,
| |
Collapse
|
23
|
Barreto IV, Pessoa FMCDP, Machado CB, Pantoja LDC, Ribeiro RM, Lopes GS, Amaral de Moraes ME, de Moraes Filho MO, de Souza LEB, Burbano RMR, Khayat AS, Moreira-Nunes CA. Leukemic Stem Cell: A Mini-Review on Clinical Perspectives. Front Oncol 2022; 12:931050. [PMID: 35814466 PMCID: PMC9270022 DOI: 10.3389/fonc.2022.931050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are known for their ability to proliferate and self-renew, thus being responsible for sustaining the hematopoietic system and residing in the bone marrow (BM). Leukemic stem cells (LSCs) are recognized by their stemness features such as drug resistance, self-renewal, and undifferentiated state. LSCs are also present in BM, being found in only 0.1%, approximately. This makes their identification and even their differentiation difficult since, despite the mutations, they are cells that still have many similarities with HSCs. Although the common characteristics, LSCs are heterogeneous cells and have different phenotypic characteristics, genetic mutations, and metabolic alterations. This whole set of alterations enables the cell to initiate the process of carcinogenesis, in addition to conferring drug resistance and providing relapses. The study of LSCs has been evolving and its application can help patients, where through its count as a biomarker, it can indicate a prognostic factor and reveal treatment results. The selection of a target to LSC therapy is fundamental. Ideally, the target chosen should be highly expressed by LSCs, highly selective, absence of expression on other cells, in particular HSC, and preferentially expressed by high numbers of patients. In view of the large number of similarities between LSCs and HSCs, it is not surprising that current treatment approaches are limited. In this mini review we seek to describe the immunophenotypic characteristics and mechanisms of resistance presented by LSCs, also approaching possible alternatives for the treatment of patients.
Collapse
Affiliation(s)
- Igor Valentim Barreto
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Flávia Melo Cunha de Pinho Pessoa
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Caio Bezerra Machado
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Laudreísa da Costa Pantoja
- Department of Pediatrics, Octávio Lobo Children’s Hospital, Belém, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
| | | | | | - Maria Elisabete Amaral de Moraes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Manoel Odorico de Moraes Filho
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | | | | | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
| | - Caroline Aquino Moreira-Nunes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
- Ceará State University, Northeast Biotechnology Network (RENORBIO), Fortaleza, Brazil
- *Correspondence: Caroline Aquino Moreira-Nunes,
| |
Collapse
|
24
|
Kreutmair S, Lippert LJ, Klingeberg C, Albers-Leischner C, Yacob S, Shlyakhto V, Mueller T, Mueller-Rudorf A, Yu C, Gorantla SP, Miething C, Duyster J, Illert AL. NIPA (Nuclear Interaction Partner of ALK) Is Crucial for Effective NPM-ALK Mediated Lymphomagenesis. Front Oncol 2022; 12:875117. [PMID: 35646639 PMCID: PMC9137267 DOI: 10.3389/fonc.2022.875117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
The NPM-ALK fusion kinase is expressed in 60% of systemic anaplastic large-cell lymphomas (ALCL). A Nuclear Interaction Partner of ALK (NIPA) was identified as a binding partner of NPM-ALK. To identify the precise role of NIPA for NPM-ALK-driven lymphomagenesis, we investigated various NPM-ALK+ cell lines and mouse models. Nipa deletion in primary mouse embryonic fibroblasts resulted in reduced transformation ability and colony formation upon NPM-ALK expression. Downregulating NIPA in murine NPM-ALK+ Ba/F3 and human ALCL cells decreased their proliferation ability and demonstrated synergistic effects of ALK inhibition and NIPA knockdown. Comprehensive in vivo analyses using short- and long-latency transplantation mouse models with NPM-ALK+ bone marrow (BM) revealed that Nipa deletion inhibited NPM-ALK-induced tumorigenesis with prolonged survival and reduced spleen colonies. To avoid off-target effects, we combined Nipa deletion and NPM-ALK expression exclusively in T cells using a lineage-restricted murine ALCL-like model resembling human disease: control mice died from neoplastic T-cell infiltration, whereas mice transplanted with Lck-CreTG/wtNipaflox/flox NPM-ALK+ BM showed significantly prolonged survival. Immunophenotypic analyses indicated a characteristic ALCL-like phenotype in all recipients but revealed fewer “stem-cell-like” features of Nipa-deficient lymphomas compared to controls. Our results identify NIPA as a crucial player in effective NPM-ALK-driven ALCL-like disease in clinically relevant murine and cell-based models.
Collapse
Affiliation(s)
- Stefanie Kreutmair
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Lena Johanna Lippert
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cathrin Klingeberg
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Corinna Albers-Leischner
- Department of Hematology, Oncology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Salome Yacob
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Valeria Shlyakhto
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tony Mueller
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department I of Internal Medicine, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Alina Mueller-Rudorf
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Chuanjiang Yu
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sivahari Prasad Gorantla
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Hematology and Oncology, Medical Center, University of Schleswig-Holstein, Lübeck, Germany
| | - Cornelius Miething
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Justus Duyster
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Anna Lena Illert
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
- *Correspondence: Anna Lena Illert,
| |
Collapse
|
25
|
Bolandi SM, Pakjoo M, Beigi P, Kiani M, Allahgholipour A, Goudarzi N, Khorashad JS, Eiring AM. A Role for the Bone Marrow Microenvironment in Drug Resistance of Acute Myeloid Leukemia. Cells 2021; 10:2833. [PMID: 34831055 PMCID: PMC8616250 DOI: 10.3390/cells10112833] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis and remarkable resistance to chemotherapeutic agents. Understanding resistance mechanisms against currently available drugs helps to recognize the therapeutic obstacles. Various mechanisms of resistance to chemotherapy or targeted inhibitors have been described for AML cells, including a role for the bone marrow niche in both the initiation and persistence of the disease, and in drug resistance of the leukemic stem cell (LSC) population. The BM niche supports LSC survival through direct and indirect interactions among the stromal cells, hematopoietic stem/progenitor cells, and leukemic cells. Additionally, the BM niche mediates changes in metabolic and signal pathway activation due to the acquisition of new mutations or selection and expansion of a minor clone. This review briefly discusses the role of the BM microenvironment and metabolic pathways in resistance to therapy, as discovered through AML clinical studies or cell line and animal models.
Collapse
Affiliation(s)
- Seyed Mohammadreza Bolandi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Peyman Beigi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Mohammad Kiani
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Ali Allahgholipour
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Negar Goudarzi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Hammersmith Hospital, Imperial College London, London W12 0HS, UK;
| | - Anna M. Eiring
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| |
Collapse
|
26
|
Zhao G, Wang Q, Li S, Wang X. Resistance to Hypomethylating Agents in Myelodysplastic Syndrome and Acute Myeloid Leukemia From Clinical Data and Molecular Mechanism. Front Oncol 2021; 11:706030. [PMID: 34650913 PMCID: PMC8505973 DOI: 10.3389/fonc.2021.706030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
The nucleoside analogs decitabine (5-AZA-dC) and azacitidine (5-AZA) have been developed as targeted therapies to reverse DNA methylation in different cancer types, and they significantly improve the survival of patients who are not suitable for traditional intensive chemotherapies or other treatment regimens. However, approximately 50% of patients have a response to hypomethylating agents (HMAs), and many patients have no response originally or in the process of treatment. Even though new combination regimens have been tested to overcome the resistance to 5-AZA-dC or 5-AZA, only a small proportion of patients benefited from these strategies, and the outcome was very poor. However, the mechanisms of the resistance remain unknown. Some studies only partially described management after failure and the mechanisms of resistance. Herein, we will review the clinical and molecular signatures of the HMA response, alternative treatment after failure, and the causes of resistance in hematological malignancies.
Collapse
Affiliation(s)
| | | | | | - Xiaoqin Wang
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Atkinson SP. A preview of select articles. Stem Cells 2021. [DOI: 10.1002/stem.3384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Hao T, Zhang C, Wang Z, Buck A, Vonderfecht SL, Ermel R, Kim Y, Chen W. An aging mouse model of human chronic myeloid leukemia. Oncogene 2021; 40:3152-3163. [PMID: 33824471 PMCID: PMC8087641 DOI: 10.1038/s41388-021-01770-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/09/2021] [Accepted: 03/23/2021] [Indexed: 11/22/2022]
Abstract
Chronic myeloid leukemia (CML) is an age-dependent blood malignancy. Like many other age-dependent human diseases, laboratory animal research of CML uses young mice that do not factor in the influence of aging. To understand how aging may impact animal modeling of human age-dependent diseases, we established the first aging mouse model of human CML in BALB/c mice in the advanced age defined by 75% survival. This model was developed by noncytotoxic depletion of bone marrow lineage-positive cells followed by BCR-ABL retroviral transduction and transplantation. CML developed in aging mice shared many similarities to that in young mice, but had increased incidence of anemia that is often seen in human CML. Importantly, we showed that aging of both donor hematopoietic stem cells and recipient bone marrow niche impacted BCR-ABL mediated leukemogenesis and leukemia spectrum. Optimal CML induction relied on age-matching for donors and recipients, and cross-transplantation between young and old mice produced a mixture of different leukemia. Therefore, our model provides initial evidence of the feasibility and merit of CML modeling in aging mice and offers a new tool for future studies of CML stem cell drug resistance and therapeutic intervention in which aging would be taken into consideration as an influencing factor.
Collapse
Affiliation(s)
- Taisen Hao
- Department of Cancer Biology, The Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Chunxiao Zhang
- Department of Cancer Biology, The Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Zhiqiang Wang
- Department of Cancer Biology, The Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Alison Buck
- Eugene and Ruth Roberts Summer Student Academy of City of Hope, Duarte, CA, USA
| | | | - Richard Ermel
- Center for Comparative Medicine, The Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Young Kim
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - WenYong Chen
- Department of Cancer Biology, The Beckman Research Institute of City of Hope, Duarte, CA, USA.
| |
Collapse
|
29
|
Han H, Byun JM, Shin DY, Yoon SS, Koh Y, Hong J, Kim I, Lee C, Yoo H, Yun H, Kim MJ, Cho SI, Seong MW, Park SS. Leukemic stem cell phenotype is associated with mutational profile in acute myeloid leukemia. Korean J Intern Med 2021; 36:401-412. [PMID: 32811132 PMCID: PMC7969060 DOI: 10.3904/kjim.2020.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIMS Understanding leukemic stem cell (LSC) is important for acute myeloid leukemia (AML) treatment. However, association of LSC with patient prognosis and genetic information in AML patients is unclear. METHODS Here we investigated the associations between genetic information and the various LSC phenotypes, namely multipotent progenitor (MPP)-like, lymphoid primed multipotent progenitor (LMPP)-like and granulocyte-macrophage progenitors (GMP)-like LSC in 52 AML patients. RESULTS In secondary AML patients, MPP-like LSC was significantly higher than de novo AML (p = 0.0037). The proportion of MPP-like LSC was especially high in post-myeloproliferative neoplasm AML (p = 0.0485). There was no correlation between age and LSC phenotype. Mutations of KRAS and NRAS were observed in MPP-like LSC dominant patients, TP53 and ASXL1 mutations in LMPP-like LSC dominant patients, and CEBPA, DNMT3A and IDH1 mutations in GMP-like LSC dominant patients. Furthermore, KRAS mutation was significantly associated with MPP-like LSC expression (p = 0.0540), and TP53 mutation with LMPP-like LSC expression (p = 0.0276). When the patients were separated according to the combined risk including next generation sequencing data, the poorer the prognosis, the higher the LMPP-like LSC expression (p = 0.0052). This suggests that the dominant phenotype of LSC is one of the important factors in predicting the prognosis and treatment of AML. CONCLUSION LSC phenotype in AML is closely associated with the recurrent mutations which has prognostic implication. Further research to confirm the meaning of LSC phenotype in the context of genetic aberration is warranted.
Collapse
Affiliation(s)
- Heejoo Han
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Ja Min Byun
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dong-Yeop Shin
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Correspondence to Dong-Yeop Shin, M.D. Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea Tel: +82-2-2072-2228 Fax: +82-2-762-9662 E-mail:
| | - Sung-Soo Yoon
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Youngil Koh
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Junshik Hong
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Inho Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Chansup Lee
- Cancer Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hyeonjoo Yoo
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hongseok Yun
- Center for Precision Medicine, Seoul National University Hospital, Seoul, Korea
| | - Man Jin Kim
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sung Im Cho
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Moon-Woo Seong
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sung Sup Park
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
30
|
Deak D, Gorcea-Andronic N, Sas V, Teodorescu P, Constantinescu C, Iluta S, Pasca S, Hotea I, Turcas C, Moisoiu V, Zimta AA, Galdean S, Steinheber J, Rus I, Rauch S, Richlitzki C, Munteanu R, Jurj A, Petrushev B, Selicean C, Marian M, Soritau O, Andries A, Roman A, Dima D, Tanase A, Sigurjonsson O, Tomuleasa C. A narrative review of central nervous system involvement in acute leukemias. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:68. [PMID: 33553361 PMCID: PMC7859772 DOI: 10.21037/atm-20-3140] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acute leukemias (both myeloid and lymphoblastic) are a group of diseases for which each year more successful therapies are implemented. However, in a subset of cases the overall survival (OS) is still exceptionally low due to the infiltration of leukemic cells in the central nervous system (CNS) and the subsequent formation of brain tumors. The CNS involvement is more common in acute lymphocytic leukemia (ALL), than in adult acute myeloid leukemia (AML), although the rates for the second case might be underestimated. The main reasons for CNS invasion are related to the expression of specific adhesion molecules (VLA-4, ICAM-1, VCAM, L-selectin, PECAM-1, CD18, LFA-1, CD58, CD44, CXCL12) by a subpopulation of leukemic cells, called “sticky cells” which have the ability to interact and adhere to endothelial cells. Moreover, the microenvironment becomes hypoxic and together with secretion of VEGF-A by ALL or AML cells the permeability of vasculature in the bone marrow increases, coupled with the disruption of blood brain barrier. There is a single subpopulation of leukemia cells, called leukemia stem cells (LSCs) that is able to resist in the new microenvironment due to its high adaptability. The LCSs enter into the arachnoid, migrate, and intensively proliferate in cerebrospinal fluid (CSF) and consequently infiltrate perivascular spaces and brain parenchyma. Moreover, the CNS is an immune privileged site that also protects leukemic cells from chemotherapy. CD56/NCAM is the most important surface molecule often overexpressed by leukemic stem cells that offers them the ability to infiltrate in the CNS. Although asymptomatic or with unspecific symptoms, CNS leukemia should be assessed in both AML/ALL patients, through a combination of flow cytometry and cytological analysis of CSF. Intrathecal therapy (ITT) is a preventive measure for CNS involvement in AML and ALL, still much research is needed in finding the appropriate target that would dramatically lower CNS involvement in acute leukemia.
Collapse
Affiliation(s)
- Dalma Deak
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Nicolae Gorcea-Andronic
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Valentina Sas
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Pediatrics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Intensive Care Unit, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sabina Iluta
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sergiu Pasca
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ionut Hotea
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Turcas
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Vlad Moisoiu
- Department of Neurosurgery, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Simona Galdean
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Jakob Steinheber
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Rus
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sebastian Rauch
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cedric Richlitzki
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bobe Petrushev
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Selicean
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Mirela Marian
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Olga Soritau
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alexandra Andries
- Department of Radiology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Andrei Roman
- Department of Radiology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Radiology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
31
|
Abstract
Defined by its potential for self-renewal, differentiation and tumorigenicity, cancer stem cells (CSCs) are considered responsible for drug resistance and relapse. To understand the behavior of CSC, the effects of the microenvironment in each tissue are a matter of great concerns for scientists in cancer biology. However, there are many complicated obstacles in the mimicking the microenvironment of CSCs even with current advanced technology. In this context, novel biomaterials have widely been assessed as in vitro platforms for their ability to mimic cancer microenvironment. These efforts should be successful to identify and characterize various CSCs specific in each type of cancer. Therefore, extracellular matrix scaffolds made of biomaterial will modulate the interactions and facilitate the investigation of CSC associated with biological phenomena simplifying the complexity of the microenvironment. In this review, we summarize latest advances in biomaterial scaffolds, which are exploited to mimic CSC microenvironment, and their chemical and biological requirements with discussion. The discussion includes the possible effects on both cells in tumors and microenvironment to propose what the critical factors are in controlling the CSC microenvironment focusing the future investigation. Our insights on their availability in drug screening will also follow the discussion.
Collapse
|
32
|
Arnone M, Konantz M, Hanns P, Paczulla Stanger AM, Bertels S, Godavarthy PS, Christopeit M, Lengerke C. Acute Myeloid Leukemia Stem Cells: The Challenges of Phenotypic Heterogeneity. Cancers (Basel) 2020; 12:E3742. [PMID: 33322769 PMCID: PMC7764578 DOI: 10.3390/cancers12123742] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 02/08/2023] Open
Abstract
Patients suffering from acute myeloid leukemia (AML) show highly heterogeneous clinical outcomes. Next to variabilities in patient-specific parameters influencing treatment decisions and outcome, this is due to differences in AML biology. In fact, different genetic drivers may transform variable cells of origin and co-exist with additional genetic lesions (e.g., as observed in clonal hematopoiesis) in a variety of leukemic (sub)clones. Moreover, AML cells are hierarchically organized and contain subpopulations of more immature cells called leukemic stem cells (LSC), which on the cellular level constitute the driver of the disease and may evolve during therapy. This genetic and hierarchical complexity results in a pronounced phenotypic variability, which is observed among AML cells of different patients as well as among the leukemic blasts of individual patients, at diagnosis and during the course of the disease. Here, we review the current knowledge on the heterogeneous landscape of AML surface markers with particular focus on those identifying LSC, and discuss why identification and targeting of this important cellular subpopulation in AML remains challenging.
Collapse
Affiliation(s)
- Marlon Arnone
- Department of Biomedicine, University of Basel and University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (M.A.); (M.K.); (P.H.)
| | - Martina Konantz
- Department of Biomedicine, University of Basel and University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (M.A.); (M.K.); (P.H.)
| | - Pauline Hanns
- Department of Biomedicine, University of Basel and University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (M.A.); (M.K.); (P.H.)
| | - Anna M. Paczulla Stanger
- Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, Department for Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.M.P.S.); (S.B.); (P.S.G.); (M.C.)
| | - Sarah Bertels
- Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, Department for Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.M.P.S.); (S.B.); (P.S.G.); (M.C.)
| | - Parimala Sonika Godavarthy
- Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, Department for Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.M.P.S.); (S.B.); (P.S.G.); (M.C.)
| | - Maximilian Christopeit
- Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, Department for Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.M.P.S.); (S.B.); (P.S.G.); (M.C.)
| | - Claudia Lengerke
- Department of Biomedicine, University of Basel and University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (M.A.); (M.K.); (P.H.)
- Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, Department for Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.M.P.S.); (S.B.); (P.S.G.); (M.C.)
| |
Collapse
|
33
|
Rossi F, Noren H, Jove R, Beljanski V, Grinnemo KH. Differences and similarities between cancer and somatic stem cells: therapeutic implications. Stem Cell Res Ther 2020; 11:489. [PMID: 33208173 PMCID: PMC7672862 DOI: 10.1186/s13287-020-02018-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Over the last decades, the cancer survival rate has increased due to personalized therapies, the discovery of targeted therapeutics and novel biological agents, and the application of palliative treatments. Despite these advances, tumor resistance to chemotherapy and radiation and rapid progression to metastatic disease are still seen in many patients. Evidence has shown that cancer stem cells (CSCs), a sub-population of cells that share many common characteristics with somatic stem cells (SSCs), contribute to this therapeutic failure. The most critical properties of CSCs are their self-renewal ability and their capacity for differentiation into heterogeneous populations of cancer cells. Although CSCs only constitute a low percentage of the total tumor mass, these cells can regrow the tumor mass on their own. Initially identified in leukemia, CSCs have subsequently been found in cancers of the breast, the colon, the pancreas, and the brain. Common genetic and phenotypic features found in both SSCs and CSCs, including upregulated signaling pathways such as Notch, Wnt, Hedgehog, and TGF-β. These pathways play fundamental roles in the development as well as in the control of cell survival and cell fate and are relevant to therapeutic targeting of CSCs. The differences in the expression of membrane proteins and exosome-delivered microRNAs between SSCs and CSCs are also important to specifically target the stem cells of the cancer. Further research efforts should be directed toward elucidation of the fundamental differences between SSCs and CSCs to improve existing therapies and generate new clinically relevant cancer treatments.
Collapse
Affiliation(s)
- Fiorella Rossi
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA
| | - Hunter Noren
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA
| | - Richard Jove
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA
| | - Vladimir Beljanski
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA.
| | - Karl-Henrik Grinnemo
- NSU Cell Therapy Institute, Nova Southeastern University, 3301 College Ave, 3200 South University Drive, Fort Lauderdale, FL, 33328, USA. .,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden. .,Department of Surgical Sciences, Division of Cardiothoracic Surgery and Anaesthesiology, Uppsala University, Akademiska University Hospital, Akademiska sjukhuset, ingång 50, 4 tr, 751 85, Uppsala, Sweden.
| |
Collapse
|
34
|
Warrier S, Patil M, Bhansali S, Varier L, Sethi G. Designing precision medicine panels for drug refractory cancers targeting cancer stemness traits. Biochim Biophys Acta Rev Cancer 2020; 1875:188475. [PMID: 33188876 DOI: 10.1016/j.bbcan.2020.188475] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
Cancer is one amongst the major causes of death today and cancer biology is one of the most well researched fields in medicine. The driving force behind cancer is considered to be a minor subpopulation of cells, the cancer stem cells (CSCs). Similar to other stem cells, these cells are self-renewing and proliferating but CSCs are also difficult to target by chemo- or radio-therapies. Cancer stem cells are known to be present in most of the cancer subgroups such as carcinoma, sarcoma, myeloma, leukemia, lymphomas and mixed cancer types. There is a wide gamut of factors attributed to the stemness of cancers, ranging from dysregulated signaling pathways, and activation of enzymes aiding immune evasion, to conducive tumor microenvironment, to name a few. The defining outcome of the increased presence of CSCs is tumor metastasis and relapse. Predictive medicine approach based on the plethora of CSC markers would be a move towards precision medicine to specifically identify CSC-rich tumors. In this review, we discuss the cancer subtypes and the role of different CSC specific markers in these varying subtypes. We also categorize the CSC markers based their defining trait contributing to stemness. This review thus provides a comprehensive approach to catalogue a predictive set of markers to identify the resistant and refractory cancer stem cell population within different tumor subtypes, so as to facilitate better prognosis and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India; Cuor Stem Cellutions Pvt Ltd, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India.
| | - Manasi Patil
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | - Sanyukta Bhansali
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | | | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117 600, Singapore
| |
Collapse
|
35
|
Lee HY, Hong IS. Targeting Liver Cancer Stem Cells: An Alternative Therapeutic Approach for Liver Cancer. Cancers (Basel) 2020; 12:cancers12102746. [PMID: 32987767 PMCID: PMC7598600 DOI: 10.3390/cancers12102746] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
The first report of cancer stem cell (CSC) from Bruce et al. has demonstrated the relatively rare population of stem-like cells in acute myeloid leukemia (AML). The discovery of leukemic CSCs prompted further identification of CSCs in multiple types of solid tumor. Recently, extensive research has attempted to identity CSCs in multiple types of solid tumors in the brain, colon, head and neck, liver, and lung. Based on these studies, we hypothesize that the initiation and progression of most malignant tumors rely largely on the CSC population. Recent studies indicated that stem cell-related markers or signaling pathways, such as aldehyde dehydrogenase (ALDH), CD133, epithelial cell adhesion molecule (EpCAM), Wnt/β-catenin signaling, and Notch signaling, contribute to the initiation and progression of various liver cancer types. Importantly, CSCs are markedly resistant to conventional therapeutic approaches and current targeted therapeutics. Therefore, it is believed that selectively targeting specific markers and/or signaling pathways of hepatic CSCs is an effective therapeutic strategy for treating chemotherapy-resistant liver cancer. Here, we provide an overview of the current knowledge on the hepatic CSC hypothesis and discuss the specific surface markers and critical signaling pathways involved in the development and maintenance of hepatic CSC subpopulations.
Collapse
Affiliation(s)
- Hwa-Yong Lee
- Department of Biomedical Science, Jungwon University, 85 Goesan-eup, Munmu-ro, Goesan-gun, Chungcheongbuk-do 367700, Korea;
| | - In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406840, Korea
- Correspondence: ; Tel.: +82-32-899-6315; Fax: +82-32-899-6350
| |
Collapse
|
36
|
Darici S, Alkhaldi H, Horne G, Jørgensen HG, Marmiroli S, Huang X. Targeting PI3K/Akt/mTOR in AML: Rationale and Clinical Evidence. J Clin Med 2020; 9:E2934. [PMID: 32932888 PMCID: PMC7563273 DOI: 10.3390/jcm9092934] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematopoietic malignancy characterized by excessive proliferation and accumulation of immature myeloid blasts in the bone marrow. AML has a very poor 5-year survival rate of just 16% in the UK; hence, more efficacious, tolerable, and targeted therapy is required. Persistent leukemia stem cell (LSC) populations underlie patient relapse and development of resistance to therapy. Identification of critical oncogenic signaling pathways in AML LSC may provide new avenues for novel therapeutic strategies. The phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling pathway, is often hyperactivated in AML, required to sustain the oncogenic potential of LSCs. Growing evidence suggests that targeting key components of this pathway may represent an effective treatment to kill AML LSCs. Despite this, accruing significant body of scientific knowledge, PI3K/Akt/mTOR inhibitors have not translated into clinical practice. In this article, we review the laboratory-based evidence of the critical role of PI3K/Akt/mTOR pathway in AML, and outcomes from current clinical studies using PI3K/Akt/mTOR inhibitors. Based on these results, we discuss the putative mechanisms of resistance to PI3K/Akt/mTOR inhibition, offering rationale for potential candidate combination therapies incorporating PI3K/Akt/mTOR inhibitors for precision medicine in AML.
Collapse
Affiliation(s)
- Salihanur Darici
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Hazem Alkhaldi
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Gillian Horne
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Heather G. Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Sandra Marmiroli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| |
Collapse
|
37
|
Wei Y, Li H, Qu Q. miR-484 suppresses endocrine therapy-resistant cells by inhibiting KLF4-induced cancer stem cells in estrogen receptor-positive cancers. Breast Cancer 2020; 28:175-186. [PMID: 32865695 DOI: 10.1007/s12282-020-01152-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/07/2020] [Indexed: 12/26/2022]
Abstract
Endocrine therapy (mainly anti-estrogen therapy) is the mainstay of treatment for estrogen receptor (ER) positive breast cancer (BCa). However, approximately one-third of BCa patients who receive endocrine therapy may develop resistance. The detailed mechanism is still unclear. MCF7 and T-47D cells were treated with ERα antagonist tamoxifen for 2 months until they became tamoxifen-resistant. qPCR was used to detect the stem markers like CD44, OCT4 and SOX2. Flow cytometry and sphere formation were performed to test the stemness. Cell growth and invasiveness were measured by MTS assay, xenograft mouse model, and invasion assay. We found that tamoxifen resistant BCa cells acquired certain malignant phenotypes, such as higher expression of KLF4, stemness and enhanced invasiveness. Furthermore, miR-484 was found to act as a tumor suppressor and directly downregulated KLF4. KLF4-induced cancer stem cell (CSCs) contributes to anti-ER therapy resistant and is a potential target in endocrine therapy-resistant cancers.
Collapse
Affiliation(s)
- Yulei Wei
- Department of Gynecology and Obstetrics, Tianjin First Central Hospital, 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
| | - Hong Li
- Department of Gynecology and Obstetrics, Tianjin First Central Hospital, 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
| | - Quanxin Qu
- Department of Gynecology and Obstetrics, Tianjin First Central Hospital, 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China.
| |
Collapse
|
38
|
Kapadia BB, Gartenhaus RB. DUBbing Down Translation: The Functional Interaction of Deubiquitinases with the Translational Machinery. Mol Cancer Ther 2020; 18:1475-1483. [PMID: 31481479 DOI: 10.1158/1535-7163.mct-19-0307] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/12/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
Abstract
Cancer cells revamp the regulatory processes that control translation to induce tumor-specific translational programs that can adapt to a hostile microenvironment as well as withstand anticancer therapeutics. Translational initiation has been established as a common downstream effector of numerous deregulated signaling pathways that together culminate in prooncogenic expression. Other mechanisms, including ribosomal stalling and stress granule assembly, also appear to be rewired in the malignant phenotype. Therefore, better understanding of the underlying perturbations driving oncogenic translation in the transformed state will provide innovative therapeutic opportunities. This review highlights deubiquitinating enzymes that are activated/dysregulated in hematologic malignancies, thereby altering the translational output and contributing to tumorigenesis.
Collapse
Affiliation(s)
| | - Ronald B Gartenhaus
- University of Maryland School of Medicine, Baltimore, Maryland. .,Veterans Administration Medical Center, Baltimore, Maryland
| |
Collapse
|
39
|
Han H, Shin DY, Kim D, Kim H, Lee C, Koh Y, Hong J, Yoon SS. Induction of leukemic stem cell differentiation by aryl hydrocarbon receptor agonist and synergy with gilteritinib in FLT3-ITD + acute myeloid leukemia. Leuk Lymphoma 2020; 61:1932-1942. [PMID: 32374198 DOI: 10.1080/10428194.2020.1747062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Leukemic stem cells (LSCs) are a major cause of treatment failure and recurrence of acute myeloid leukemia (AML). Targeting LSC is essential to developing a potential cure for patients with relapsed/refractory AML. Here we investigated the effect of aryl hydrocarbon receptor (AhR) signaling on AML stem/progenitor proportion and examined the combined effect of AhR agonist and tyrosine kinase inhibitor. The AhR agonist, 6-formylindolo[3,2-b]carbazole (FICZ), significantly decreased the LSC proportion and clonogenicity and increased differentiation markers in AML primary cells. Synergistic/additive effects of FICZ and gilteritinib, FMS-like tyrosine kinase 3 (FLT3) inhibitor, were confirmed in AML cells with FLT3-ITD. We present evidence that combination of both agents inhibits FLT3 downstream molecules and degrades clonogenicity. Collectively, our results suggest that FICZ not only compels LSC differentiation, but also enhances the efficacy of gilteritinib when combined. Clinical application of this combined approach may pave a new therapeutic strategy for patients with FLT3 mutated AML.
Collapse
Affiliation(s)
- Heejoo Han
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dongchan Kim
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyungsuk Kim
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chansup Lee
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngil Koh
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junshik Hong
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung-Soo Yoon
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
40
|
Thankamony AP, Saxena K, Murali R, Jolly MK, Nair R. Cancer Stem Cell Plasticity - A Deadly Deal. Front Mol Biosci 2020; 7:79. [PMID: 32426371 PMCID: PMC7203492 DOI: 10.3389/fmolb.2020.00079] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
Intratumoral heterogeneity is a major ongoing challenge in the effective therapeutic targeting of cancer. Accumulating evidence suggests that a fraction of cells within a tumor termed Cancer Stem Cells (CSCs) are primarily responsible for this diversity resulting in therapeutic resistance and metastasis. Adding to this complexity, recent studies have shown that there can be different subpopulations of CSCs with varying biochemical and biophysical traits resulting in varied dissemination and drug-resistance potential. Moreover, cancer cells can exhibit a high level of plasticity or the ability to dynamically switch between CSC and non-CSC states or among different subsets of CSCs. In addition, CSCs also display extensive metabolic plasticity. The molecular mechanisms underlying these different interconnected axes of plasticity has been under extensive investigation and the trans-differentiation process of Epithelial to Mesenchymal transition (EMT) has been identified as a major contributing factor. Besides genetic and epigenetic factors, CSC plasticity is also shaped by non-cell-autonomous effects such as the tumor microenvironment (TME). In this review, we discuss the latest developments in decoding mechanisms and implications of CSC plasticity in tumor progression at biochemical and biophysical levels, and the latest in silico approaches being taken for characterizing cancer cell plasticity. These efforts can help improve existing therapeutic approaches by taking into consideration the contribution of cellular plasticity/heterogeneity in enabling drug resistance.
Collapse
Affiliation(s)
- Archana P. Thankamony
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Kritika Saxena
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Reshma Murali
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Radhika Nair
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
41
|
Annageldiyev C, Tan SF, Thakur S, Dhanyamraju PK, Ramisetti SR, Bhadauria P, Schick J, Zeng Z, Sharma V, Dunton W, Dovat S, Desai D, Zheng H, Feith DJ, Loughran TP, Amin S, Sharma AK, Claxton D, Sharma A. The PI3K/AKT Pathway Inhibitor ISC-4 Induces Apoptosis and Inhibits Growth of Leukemia in Preclinical Models of Acute Myeloid Leukemia. Front Oncol 2020; 10:393. [PMID: 32296637 PMCID: PMC7140985 DOI: 10.3389/fonc.2020.00393] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia is a heterogeneous disease with a 5-year survival rate of 28.3%, and current treatment options constrained by dose-limiting toxicities. One of the key signaling pathways known to be frequently activated and dysregulated in AML is PI3K/AKT. Its dysregulation is associated with aggressive cell growth and drug resistance. We investigated the activity of Phenybutyl isoselenocyanate (ISC-4) in primary cells obtained from newly diagnosed AML patients, diverse AML cell lines, and normal cord blood cells. ISC-4 significantly inhibited survival and clonogenicity of primary human AML cells without affecting normal cells. We demonstrated that ISC-4-mediated p-Akt inhibition caused apoptosis in primary AML (CD34+) stem cells and enhanced efficacy of cytarabine. ISC-4 impeded leukemia progression with improved overall survival in a syngeneic C1498 mouse model with no obvious toxic effects on normal myelopoiesis. In U937 xenograft model, bone marrow cells exhibited significant reduction in human CD45+ cells in ISC-4 (~87%) or AraC (~89%) monotherapy groups compared to control. Notably, combination treatment suppressed the leukemic infiltration significantly higher than the single-drug treatments (~94%). Together, the present findings suggest that ISC-4 might be a promising agent for AML treatment.
Collapse
Affiliation(s)
- Charyguly Annageldiyev
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Su-Fern Tan
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Shreya Thakur
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Pavan Kumar Dhanyamraju
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Srinivasa R Ramisetti
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Preeti Bhadauria
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Jacob Schick
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Zheng Zeng
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Varun Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Wendy Dunton
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Sinisa Dovat
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Dhimant Desai
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Hong Zheng
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David J Feith
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States.,Division of Hematology and Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Thomas P Loughran
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States.,Division of Hematology and Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, United States
| | - Shantu Amin
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Arun K Sharma
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David Claxton
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Arati Sharma
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
42
|
Fathi E, Farahzadi R, Sheervalilou R, Sanaat Z, Vietor I. A general view of CD33 + leukemic stem cells and CAR-T cells as interesting targets in acute myeloblatsic leukemia therapy. Blood Res 2020; 55:10-16. [PMID: 32269970 PMCID: PMC7106116 DOI: 10.5045/br.2020.55.1.10] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/27/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
Acute myeloblastic leukemia (AML) is the most frequent acute leukemia in adulthood with very poor overall survival rates. In the past few decades, significant progresses had led to the findings of new therapeutic approaches and the better understanding of the molecular complexity of this hematologic malignancy. Leukemic stem cells (LSCs) play a key role in the initiation, progression, regression, and drug resistance of different types of leukemia. The cellular and molecular characteristics of LSCs and their mechanism in the development of leukemia had not yet been specified. Therefore, determining their cellular and molecular characteristics and creating new approaches for targeted therapy of LSCs is crucial for the future of leukemia research. For this reason, the recognition of surface maker targets on the cell surface of LSCs has attracted much attention. CD33 has been detected on blasts in most AML patients, making them an interesting target for AML therapy. Genetic engineering of T cells with chimeric antigen receptor (CAR-T cell therapy) is a novel therapeutic strategy. It extends the range of antigens available for use in adoptive T-cell immunotherapy. This review will focus on CAR-T cell approaches as well as monoclonal antibody (mAB)-based therapy, the two antibody-based therapies utilized in AML treatment.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghayeh Sheervalilou
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Zohreh Sanaat
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ilja Vietor
- Division of Cell Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
43
|
Alvarado-Ortiz E, de la Cruz-López KG, Becerril-Rico J, Sarabia-Sánchez MA, Ortiz-Sánchez E, García-Carrancá A. Mutant p53 Gain-of-Function: Role in Cancer Development, Progression, and Therapeutic Approaches. Front Cell Dev Biol 2020; 8:607670. [PMID: 33644030 PMCID: PMC7905058 DOI: 10.3389/fcell.2020.607670] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/23/2020] [Indexed: 02/05/2023] Open
Abstract
Frequent p53 mutations (mutp53) not only abolish tumor suppressor capacities but confer various gain-of-function (GOF) activities that impacts molecules and pathways now regarded as central for tumor development and progression. Although the complete impact of GOF is still far from being fully understood, the effects on proliferation, migration, metabolic reprogramming, and immune evasion, among others, certainly constitute major driving forces for human tumors harboring them. In this review we discuss major molecular mechanisms driven by mutp53 GOF. We present novel mechanistic insights on their effects over key functional molecules and processes involved in cancer. We analyze new mechanistic insights impacting processes such as immune system evasion, metabolic reprogramming, and stemness. In particular, the increased lipogenic activity through the mevalonate pathway (MVA) and the alteration of metabolic homeostasis due to interactions between mutp53 and AMP-activated protein kinase (AMPK) and Sterol regulatory element-binding protein 1 (SREBP1) that impact anabolic pathways and favor metabolic reprograming. We address, in detail, the impact of mutp53 over metabolic reprogramming and the Warburg effect observed in cancer cells as a consequence, not only of loss-of-function of p53, but rather as an effect of GOF that is crucial for the imbalance between glycolysis and oxidative phosphorylation. Additionally, transcriptional activation of new targets, resulting from interaction of mutp53 with NF-kB, HIF-1α, or SREBP1, are presented and discussed. Finally, we discuss perspectives for targeting molecules and pathways involved in chemo-resistance of tumor cells resulting from mutp53 GOF. We discuss and stress the fact that the status of p53 currently constitutes one of the most relevant criteria to understand the role of autophagy as a survival mechanism in cancer, and propose new therapeutic approaches that could promote the reduction of GOF effects exercised by mutp53 in cancer.
Collapse
Affiliation(s)
- Eduardo Alvarado-Ortiz
- Programa de Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Karen Griselda de la Cruz-López
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
- Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jared Becerril-Rico
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Miguel Angel Sarabia-Sánchez
- Programa de Posgrado en Ciencias Bioquímicas, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
| | - Alejandro García-Carrancá
- Laboratorio de Virus and Cáncer, Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
- *Correspondence: Alejandro García-Carrancá
| |
Collapse
|
44
|
Triaca V, Carito V, Fico E, Rosso P, Fiore M, Ralli M, Lambiase A, Greco A, Tirassa P. Cancer stem cells-driven tumor growth and immune escape: the Janus face of neurotrophins. Aging (Albany NY) 2019; 11:11770-11792. [PMID: 31812953 PMCID: PMC6932930 DOI: 10.18632/aging.102499] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/17/2019] [Indexed: 05/12/2023]
Abstract
Cancer Stem Cells (CSCs) are self-renewing cancer cells responsible for expansion of the malignant mass in a dynamic process shaping the tumor microenvironment. CSCs may hijack the host immune surveillance resulting in typically aggressive tumors with poor prognosis.In this review, we focus on neurotrophic control of cellular substrates and molecular mechanisms involved in CSC-driven tumor growth as well as in host immune surveillance. Neurotrophins have been demonstrated to be key tumor promoting signaling platforms. Particularly, Nerve Growth Factor (NGF) and its specific receptor Tropomyosin related kinase A (TrkA) have been implicated in initiation and progression of many aggressive cancers. On the other hand, an active NGF pathway has been recently proven to be critical to oncogenic inflammation control and in promoting immune response against cancer, pinpointing possible pro-tumoral effects of NGF/TrkA-inhibitory therapy.A better understanding of the molecular mechanisms involved in the control of tumor growth/immunoediting is essential to identify new predictive and prognostic intervention and to design more effective therapies. Fine and timely modulation of CSCs-driven tumor growth and of peripheral lymph nodes activation by the immune system will possibly open the way to precision medicine in neurotrophic therapy and improve patient's prognosis in both TrkA- dependent and independent cancers.
Collapse
Affiliation(s)
- Viviana Triaca
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Monterotondo Scalo, Rome, Italy
| | - Valentina Carito
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Elena Fico
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Pamela Rosso
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | | | - Antonio Greco
- Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Paola Tirassa
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
45
|
Cortes JE, Dombret H, Merchant A, Tauchi T, DiRienzo CG, Sleight B, Zhang X, Leip EP, Shaik N, Bell T, Chan G, Sekeres MA. Glasdegib plus intensive/nonintensive chemotherapy in untreated acute myeloid leukemia: BRIGHT AML 1019 Phase III trials. Future Oncol 2019; 15:3531-3545. [DOI: 10.2217/fon-2019-0373] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glasdegib, an oral Hedgehog pathway inhibitor, has been associated with significantly improved survival when combined with low-dose cytarabine in patients with untreated acute myeloid leukemia (AML) who were unsuitable for intensive chemotherapy, when compared with low-dose cytarabine alone. BRIGHT AML 1019 (NCT03416179) comprises two independently powered Phase III, randomized (1:1), double-blind global trials evaluating oral glasdegib 100 mg once daily or placebo plus one of two standard chemotherapy regimens in adults with untreated AML. The intensive trial combines glasdegib/placebo with cytarabine and daunorubicin (7 + 3), while the nonintensive trial combines glasdegib/placebo with azacitidine. The primary end point of both studies is overall survival. Secondary end points include response, time to and duration of response, event-free survival, safety, patient-reported outcomes and pharmacokinetics. Trial registration number: ClinicalTrials.gov identifier: NCT03416179
Collapse
Affiliation(s)
- Jorge E Cortes
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Hervé Dombret
- Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris Institut Universitaire d`Hematologie Hopital Saint Louis, Paris, France
| | - Akil Merchant
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Tetsuzo Tauchi
- Department of Hematology, Shin-Yurigaoka General Hospital, Kawasaki, Japan
| | | | | | - Xiaoxi Zhang
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | - Eric P Leip
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | - Naveed Shaik
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | - Timothy Bell
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | - Geoffrey Chan
- Pfizer Oncology, Pfizer Inc., New York, NY 10017, USA
| | | |
Collapse
|
46
|
Bellina M. [Leukemic stem cells: targeting quiescence to optimize therapy]. Med Sci (Paris) 2019; 35:705-708. [PMID: 31532386 DOI: 10.1051/medsci/2019140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Mélanie Bellina
- Master Biologie Moléculaire et Cellulaire-M2 Parcours Génopath, Université Claude Bernard Lyon 1, Université de Lyon, France
| |
Collapse
|
47
|
Epigenetic Abnormalities in Acute Myeloid Leukemia and Leukemia Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31338820 DOI: 10.1007/978-981-13-7342-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2023]
Abstract
Recently advances in cancer genomics revealed the unexpected high frequencies of epigenetic abnormalities in human acute myeloid leukemia (AML). Accumulating data suggest that these leukemia-associated epigenetic factors play critical roles in both normal hematopoietic stem cells (HSCs) and leukemia stem cells (LSCs). In turn, these abnormalities result in susceptibilities of LSC and related diseases to epigenetic inhibitors. In this chapter, we will focus on the mutations of epigenetic factors in AML, their functional roles and mechanisms in normal hematopoiesis and leukemia genesis, especially in LSC, and potential treatment opportunities specifically for AML with epigenetic dysregulations.
Collapse
|
48
|
Ahmed ESA, Ahmed NH, Medhat AM, Said UZ, Rashed LA, Abdel Ghaffar ARB. Mesenchymal stem cells targeting PI3K/AKT pathway in leukemic model. Tumour Biol 2019; 41:1010428319846803. [PMID: 31018830 DOI: 10.1177/1010428319846803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells have therapeutic properties that are related to their potentials for trans-differentiation, immunomodulation, anti-inflammatory, inhibitory effect on tumor proliferation, and induction of apoptosis. This study was performed to analyze the role of mesenchymal stem cells as an alternative for cellular signaling growth factors involved in the pathogenesis of leukemogenesis in rats. Treatment of rats with 7,12-dimethyl benz [a] anthracene induced leukemogenesis appeared as a significant decrease in hematological parameters with concomitant significant increase in bone marrow oxidative and inflammatory indices (transforming growth factor beta and interleukin-6) in comparison with normal groups. On the contrary, Western immunoblotting showed a significant increase in the signaling growth factors: PI3K, AKT, mTOR proteins and a significant decrease in PTEN in 7,12-dimethyl benz [a] anthracene-treated group. In addition, a significant increase in the transcript levels of B cell lymphoma-2 protein gene in the 7,12-dimethyl benz [a] anthracene group, while that of C-X-C motif chemokine receptor-4 and B cell lymphoma-2 protein associated x-protein were significantly downregulated compared to controls. Meanwhile, therapeutic mesenchymal stem cells treatment predict a significant improvement versus 7,12-dimethyl benz [a] anthracene group through the modulation of growth factors that confront bone marrow dysplasia. In the same direction treatment of 7,12-dimethyl benz [a] anthracene group with mesenchymal stem cells, it induced apoptosis and increased the homing efficacy to bone marrow. In conclusion, mesenchymal stem cells improve hematopoiesis and alleviate inflammation, and modulated PI3K/AKT signaling pathway contributed to experimental leukemogenesis.
Collapse
Affiliation(s)
- Esraa S A Ahmed
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Neamat H Ahmed
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Amina M Medhat
- 2 Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ussama Z Said
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Laila A Rashed
- 3 Biochemistry & Molecular Biology Department, Medicine Faculty, Cairo University, Cairo, Egypt
| | | |
Collapse
|
49
|
Gacha-Garay MJ, Niño-Joya AF, Bolaños NI, Abenoza L, Quintero G, Ibarra H, Gonzalez JM, Akle V, Garavito-Aguilar ZV. Pilot Study of an Integrative New Tool for Studying Clinical Outcome Discrimination in Acute Leukemia. Front Oncol 2019; 9:245. [PMID: 31024847 PMCID: PMC6465644 DOI: 10.3389/fonc.2019.00245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/18/2019] [Indexed: 01/04/2023] Open
Abstract
Acute leukemia is a heterogeneous set of diseases affecting children and adults. Current prognostic factors are not accurate predictors of the clinical outcome of adult patients and the stratification of risk groups remains insufficient. For that reason, this study proposes a multifactorial analysis which integrates clinical parameters, ex vivo tumor characterization and behavioral in vivo analysis in zebrafish. This model represents a new approach to understand leukemic primary cells behavior and features associated with aggressiveness and metastatic potential. Xenotransplantation of primary samples from patients newly diagnosed with acute leukemia in zebrafish embryos at 48 hpf was used to asses survival rate, dissemination pattern, and metastatic potential. Seven samples from young adults classified in adverse, favorable or intermediate risk group were characterized. Tumor heterogeneity defined by Leukemic stem cell (LSC) proportion, was performed by metabolic and cell membrane biomarkers characterization. Thus, our work combines all these parameters with a robust quantification strategy that provides important information about leukemia biology, their relationship with specific niches and the existent inter and intra-tumor heterogeneity in acute leukemia. In regard to prognostic factors, leukemic stem cell proportion and Patient-derived xenografts (PDX) migration into zebrafish were the variables with highest weights for the prediction analysis. Higher ALDH activity, less differentiated cells and a broader and random migration pattern are related with worse clinical outcome after induction chemotherapy. This model also recapitulates multiple aspects of human acute leukemia and therefore is a promising tool to be employed not only for preclinical studies but also supposes a new tool with a higher resolution compared to traditional methods for an accurate stratification of patients into worse or favorable clinical outcome.
Collapse
Affiliation(s)
- María José Gacha-Garay
- Laboratory of Developmental Biology, Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - Andrés Felipe Niño-Joya
- Laboratory of Developmental Biology, Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - Natalia I Bolaños
- Biomedical Sciences Group, School of Medicine, Universidad de los Andes, Bogotá, Colombia
| | - Lina Abenoza
- Department of Oncology, Fundación Santa Fe de Bogotá, Bogotá, Colombia
| | | | - Humberto Ibarra
- Microscopy Core, Vice-Presidency of Research, Universidad de los Andes, Bogotá, Colombia
| | - John M Gonzalez
- Biomedical Sciences Group, School of Medicine, Universidad de los Andes, Bogotá, Colombia
| | - Verónica Akle
- Laboratory of Neuroscience and Circadian Rhythms, School of Medicine, Universidad de los Andes, Bogotá, Colombia
| | - Zayra V Garavito-Aguilar
- Laboratory of Developmental Biology, Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
50
|
The roles of circular RNAs in human development and diseases. Biomed Pharmacother 2019; 111:198-208. [DOI: 10.1016/j.biopha.2018.12.052] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 12/20/2022] Open
|