1
|
Yang X, Chi L, Qiao M, Huang A, Wu H, Chen S, Fan J, Lin X, Chen J. The effect of C-reactive protein and interleukin-3 on mild cognitive impairment with APOE ɛ4. J Alzheimers Dis 2025:13872877251333149. [PMID: 40261294 DOI: 10.1177/13872877251333149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BackgroundThe apolipoprotein E ε4 allele (APOE ε4) and inflammation are associated with Alzheimer's disease (AD) pathology. Mild cognitive impairment (MCI) is considered the preclinical and early stage of AD. However, the comprehensive effects of APOE ε4 and inflammatory mediators on MCI patients with specific APOE ε4 genotypes remain poorly understood.ObjectiveOur study aimed to explore how different numbers of the APOE ε4 alleles affect plasma C-reactive protein (CRP) and interleukin-3 (IL-3) levels and their associations with brain structure.MethodsA total of 339 MCI patients from the Alzheimer's Disease Neuroimaging Initiative study were enrolled. We compared their plasma concentrations of CRP and IL-3, cognitive performance, and cerebrospinal fluid (CSF) AD biomarkers levels across different APOE ε4 genotypes. Structural magnetic resonance imaging was utilized to measure gray matter volume outcomes. Pearson correlation analysis was used to explore the associations between the above indicators.ResultsPlasma CRP levels increased in the APOE ε4 carriers, but IL-3 expression notably decreased, and the homozygous state is the most significant. A negative correlation between CRP and several cognitive abilities was observed only in APOE ε4 homozygotes. Additionally, a positive correlation between IL-3, cognitive scores, and CSF biomarker levels was confirmed only in APOE ε4 homozygotes. Imaging data demonstrated that the gray matter volume of the right middle frontal gyrus was associated with CRP only in APOE ε4 non-carriers.ConclusionsOur study demonstrated that peripheral inflammatory mediators' effect on cognitive function and brain structure in MCI patients differs based on their APOE ε4 allele carrier status.
Collapse
Affiliation(s)
- Xinyi Yang
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Chi
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Meizhao Qiao
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Anxing Huang
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Huimin Wu
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Shanshan Chen
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jia Fan
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Xingjian Lin
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Chen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
2
|
Ji RJ, Cao GH, Zhao WQ, Wang MY, Gao P, Zhang YZ, Wang XB, Qiu HY, Chen DD, Tong XH, Duan M, Yin H, Zhang Y. Epitope prime editing shields hematopoietic cells from CD123 immunotherapy for acute myeloid leukemia. Cell Stem Cell 2024; 31:1650-1666.e8. [PMID: 39353428 DOI: 10.1016/j.stem.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/28/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Acute myeloid leukemia (AML) is a malignant cancer characterized by abnormal differentiation of hematopoietic stem and progenitor cells (HSPCs). While chimeric antigen receptor T (CAR-T) cell immunotherapies target AML cells, they often induce severe on-target/off-tumor toxicity by attacking normal cells expressing the same antigen. Here, we used base editors (BEs) and a prime editor (PE) to modify the epitope of CD123 on HSPCs, protecting healthy cells from CAR-T-induced cytotoxicity while maintaining their normal function. Although BE effectively edits epitopes, complex bystander products are a concern. To enhance precision, we optimized prime editing, increasing the editing efficiency from 5.9% to 78.9% in HSPCs. Epitope-modified cells were resistant to CAR-T lysis while retaining normal differentiation and function. Furthermore, BE- or PE-edited HSPCs infused into humanized mice endowed myeloid lineages with selective resistance to CAR-T immunotherapy, demonstrating a proof-of-concept strategy for treating relapsed AML.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Humans
- Interleukin-3 Receptor alpha Subunit/metabolism
- Animals
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/metabolism
- Mice
- Epitopes/immunology
- Immunotherapy/methods
- Gene Editing
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Mice, Inbred NOD
Collapse
Affiliation(s)
- Rui-Jin Ji
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Guo-Hua Cao
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Wei-Qiang Zhao
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Mu-Yao Wang
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Pan Gao
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yi-Zhou Zhang
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xue-Bin Wang
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Hou-Yuan Qiu
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Di-Di Chen
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xiao-Han Tong
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Min Duan
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Hao Yin
- Departments of Clinical Laboratory and Department of Urology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; TaiKang Centre for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan 430071, China; State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China
| | - Ying Zhang
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; TaiKang Centre for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan 430071, China; State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
3
|
Jen WY, Konopleva M, Pemmaraju N. Tagraxofusp, a first-in-class CD123-targeted agent: Five-year postapproval comprehensive review of the literature. Cancer 2024; 130:2260-2271. [PMID: 38620053 DOI: 10.1002/cncr.35315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/17/2024]
Abstract
Tagraxofusp is a first-in-class CD123-directed conjugate of an amended diphtheria toxin platform and recombinant interleukin 3. Binding and subsequent internalization of the drug result in cell death via disruption of intracellular protein synthesis. CD123 is a surface marker that is expressed in several hematological malignancies, especially blastic plasmacytoid dendritic cell neoplasm (BPDCN), where its expression is ubiquitous. A pivotal study of tagraxofusp in BPDCN resulted in its approval for the treatment of BPDCN, the first treatment approved for this indication. Since the introduction of tagraxofusp, research has focused on the management of adverse effects, combination therapy to improve outcomes in fit patients, and dosing and combination strategies to mitigate toxicities while preserving efficacy, especially among older patients. The successful targeting of CD123 in BPDCN has also encouraged research into a variety of other CD123-positive hematological neoplasms, including acute myeloid leukemia (AML), and informed the development of other novel agents targeting CD123. This review examines the clinical data leading to the development and approval of tagraxofusp in BPDCN, how it is being used in combination to improve outcomes in BPDCN and AML, and its developing role in other hematological malignancies.
Collapse
Affiliation(s)
- Wei-Ying Jen
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marina Konopleva
- Department of Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York, USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Yu J, Li J, Shen A, Liu Z, He TS. E3 ubiquitin ligase RNF128 negatively regulates the IL-3/STAT5 signaling pathway by facilitating K27-linked polyubiquitination of IL-3Rα. Cell Commun Signal 2024; 22:254. [PMID: 38702781 PMCID: PMC11067302 DOI: 10.1186/s12964-024-01636-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024] Open
Abstract
IL-3/STAT5 signaling pathway is crucial for the development and activation of immune cells, contributing to the cellular response to infections and inflammatory stimuli. Dysregulation of the IL-3/STAT5 signaling have been associated with inflammatory and autoimmune diseases characterized by inflammatory cell infiltration and organ damage. IL-3 receptor α (IL-3Rα) specifically binds to IL-3 and initiates intracellular signaling, resulting in the phosphorylation of STAT5. However, the regulatory mechanisms of IL-3Rα remain unclear. Here, we identified the E3 ubiquitin ligase RNF128 as a negative regulator of IL-3/STAT5 signaling by targeting IL-3Rα for lysosomal degradation. RNF128 was shown to selectively bind to IL-3Rα, without interacting with the common beta chain IL-3Rβ, which shares the subunit with GM-CSF. The deficiency of Rnf128 had no effect on GM-CSF-induced phosphorylation of Stat5, but it resulted in heightened Il-3-triggered activation of Stat5 and increased transcription of the Id1, Pim1, and Cd69 genes. Furthermore, we found that RNF128 promoted the K27-linked polyubiquitination of IL-3Rα in a ligase activity-dependent manner, ultimately facilitating its degradation through the lysosomal pathway. RNF128 inhibited the activation and chemotaxis of macrophages in response to LPS stimulation, thereby attenuating excessive inflammatory responses. Collectively, these results reveal that RNF128 negatively regulates the IL-3/STAT5 signaling pathway by facilitating K27-linked polyubiquitination of IL-3Rα. This study uncovers E3 ubiquitin ligase RNF128 as a novel regulator of the IL-3/STAT5 signaling pathway, providing potential molecular targets for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Jingge Yu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Blood Transfusion, Jingmen Central Hospital, Jingmen, China
| | - Jianguo Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Ao Shen
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
- School of Graduate, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhiping Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| | - Tian-Sheng He
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
5
|
Matus V, Castro-Guarda M, Cárcamo-Fierro J, Morera FJ, Zambrano A. Interleukin 3 Inhibits Glutamate-Cytotoxicity in Neuroblastoma Cell Line. Neurochem Res 2024; 49:1373-1386. [PMID: 38512424 DOI: 10.1007/s11064-024-04123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 03/23/2024]
Abstract
Interleukin 3 (IL-3) is a well-known pleiotropic cytokine that regulates the proliferation and differentiation of hematopoietic progenitor cells, triggering classical signaling pathways such as JAK/STAT, Ras/MAPK, and PI3K/Akt to carry out its functions. Interestingly, the IL-3 receptor is also expressed in non-hematopoietic cells, playing a crucial role in cell survival. Our previous research demonstrated the expression of the IL-3 receptor in neuron cells and its protective role in neurodegeneration. Glutamate, a principal neurotransmitter in the central nervous system, can induce cellular stress and lead to neurotoxicity when its extracellular concentrations surpass normal levels. This excessive glutamate presence is frequently observed in various neurological diseases. In this study, we uncover the protective role of IL-3 as an inhibitor of glutamate-induced cell death, analyzing the cytokine's signaling pathways during its protective effect. Specifically, we examined the relevance of JAK/STAT, Ras/MAPK, and PI3 K signaling pathways in the molecular mechanism triggered by IL-3. Our results show that the inhibition of JAK, ERK, and PI3 K signaling pathways, using pharmacological inhibitors, effectively blocked IL-3's protective role against glutamate-induced cell death. Additionally, our findings suggest that Bcl-2 and Bax proteins may be involved in the molecular mechanism triggered by IL-3. Our investigation into IL-3's ability to protect neuronal cells from glutamate-induced damage offers a promising therapeutic avenue with potential clinical implications for several neurological diseases characterized by glutamate neurotoxicity.
Collapse
Affiliation(s)
- Verónica Matus
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Marcos Castro-Guarda
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Joaquín Cárcamo-Fierro
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile
| | - Francisco J Morera
- Applied Biochemistry Laboratory, Escuela de Medicina Veterinaria, Facultad de Agronomía y Sistemas Naturales, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, 7820436, Santiago, Chile
| | - Angara Zambrano
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, (P. O. Box) 567, 5090000, Casilla, Valdivia, Chile.
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
6
|
Boyiadzis M, Hong CS, Yerneni S, Im A, Diergaarde B, Whiteside TL. Blast-Derived Small Extracellular Vesicles in the Plasma of Patients with Acute Myeloid Leukemia Predict Responses to Chemotherapy. Biomedicines 2023; 11:3236. [PMID: 38137457 PMCID: PMC10740822 DOI: 10.3390/biomedicines11123236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
The small extracellular vesicles (sEV) accumulating in acute myeloid leukemia (AML) patients' plasma are mixtures of vesicles produced by leukemic and non-malignant cells. sEV originating from leukemia blasts could serve as potential non-invasive biomarkers of AML response to therapy. To isolate blast-derived sEV from patients' plasma, we developed a bioprinted microarray-based immunoassay using monoclonal antibodies (mAbs) specific for leukemia-associated antigens (LAAs) and mAbs specific for a mix of tetraspanins (CD9, CD63, and CD81). We determined the proportion of LAA+ sEV relative to total plasma sEV (the LAA+/total sEV ratio) in serially collected samples of newly diagnosed AML patients prior to, during, and after chemotherapy. At AML diagnosis, the LAA+/total sEV ratio was significantly higher in patients than in healthy donors (HDs). In patients who achieved complete remission (CR) after induction chemotherapy, the LAA+/total sEV ratios significantly decreased after each chemotherapy cycle to levels seen in HDs. In contrast, the LAA+/total sEV ratios in AML patients with persistent leukemia after therapy remained elevated during and after therapy, as did the percentage of leukemic blasts in these patients' bone marrows. The LAA+/total sEV ratio emerges as a promising non-invasive biomarker of leukemia response to therapy.
Collapse
Affiliation(s)
- Michael Boyiadzis
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
| | - Chang-Sook Hong
- Department of Pathology, University of Pittsburgh School of Medicine Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | | | - Annie Im
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Brenda Diergaarde
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Theresa L. Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
7
|
Gambles MT, Yang J, Kopeček J. Multi-targeted immunotherapeutics to treat B cell malignancies. J Control Release 2023; 358:232-258. [PMID: 37121515 PMCID: PMC10330463 DOI: 10.1016/j.jconrel.2023.04.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The concept of multi-targeted immunotherapeutic systems has propelled the field of cancer immunotherapy into an exciting new era. Multi-effector molecules can be designed to engage with, and alter, the patient's immune system in a plethora of ways. The outcomes can vary from effector cell recruitment and activation upon recognition of a cancer cell, to a multipronged immune checkpoint blockade strategy disallowing evasion of the cancer cells by immune cells, or to direct cancer cell death upon engaging multiple cell surface receptors simultaneously. Here, we review the field of multi-specific immunotherapeutics implemented to treat B cell malignancies. The mechanistically diverse strategies are outlined and discussed; common B cell receptor antigen targeting strategies are outlined and summarized; and the challenges of the field are presented along with optimistic insights for the future.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
8
|
Pelosi E, Castelli G, Testa U. CD123 a Therapeutic Target for Acute Myeloid Leukemia and Blastic Plasmocytoid Dendritic Neoplasm. Int J Mol Sci 2023; 24:2718. [PMID: 36769040 PMCID: PMC9917129 DOI: 10.3390/ijms24032718] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
In spite of consistent progress at the level of basic research and of clinical treatment, acute myeloid leukemia (AML) still represents an unmet clinical need for adult and pediatric patients. To improve the outcomes of these patients, it is necessary to identify new therapeutic targets. IL3RA (CD123, alpha subunit of the interleukin 3 receptor) is a cell membrane protein overexpressed in several hematologic malignancies, including AML blastic plasmocytoid dendritic cell neoplasms (BPDCN). Given the higher expression of CD123 on leukemic cells compared to normal hematopoietic cells and its low/absent expression on normal hematopoietic stem cells, it appears as a suitable and attractive target for therapy. Various drugs targeting CD123 have been developed and evaluated at clinical level: interleukin-3 conjugated with diphtheria toxin; naked neutralizing anti-CD123 antibodies; drug-antibody conjugates; bispecific antibodies targeting both CD123 and CD3; and chimeric antigen receptor (CAR) T cells engineered to target CD123. Some of these agents have shown promising results at the clinical level, including tagraxofusp (CD123 conjugated with diphtheria toxin) for the treatment of BPDCN and IMGN632 (anti-CD123 drug-conjugate), and flotetuzumab (bispecific anti-CD123 and anti-CD3 monoclonal antibody) for the treatment of AML. However, the therapeutic efficacy of CD123-targeting treatments is still unsatisfactory and must be improved through new therapeutic strategies and combined treatments with other antileukemic drugs.
Collapse
Affiliation(s)
| | | | - Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
9
|
Xu S, Zhang M, Fang X, Hu X, Xing H, Yang Y, Meng J, Wen T, Liu J, Wang J, Wang C, Xu H. CD123 Antagonistic Peptides Assembled with Nanomicelles Act as Monotherapeutics to Combat Refractory Acute Myeloid Leukemia. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38584-38593. [PMID: 35977045 DOI: 10.1021/acsami.2c11538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. Due to the development of drug resistance to traditional chemotherapies and high relapse rate, AML still has a low survival rate and there is in an urgent need for better treatment strategies. CD123 is widely expressed by AML cells, also associated with the poor prognosis of AML. In this study, we fabricated nanomicelles loaded with a lab-designed CD123 antagonistic peptide, which were referred to as mPO-6. The antagonistic and therapeutic effects were investigated with CD123+ AML cell lines and a refractory AML mouse (AE and CKITD816V) model. Results show that mPO-6 can specifically bind to the CD123+ AML cells and inhibit the cell viability effectively. Intravenous administration of mPO-6 significantly reduces the percentage of AML cells' infiltration and prolongs the median survival of AML mice. Further, the efficiency of mPO-6 is demonstrated to interfere with the axis of CD123/IL-3 via regulating the activation of STAT5, PI3K/AKT, and NF-κB signaling pathways related to cell proliferation or apoptosis at the level of mRNA and protein in vivo and in vitro. In conclusion, the novel CD123 antagonistic peptide micelle formulation mPO-6 can significantly enhance apoptosis and prolong the survival of AML mice by effectively interfering with the axis of CD123/IL-3 and therefore is a promising therapeutic candidate for the treatment of refractory AML.
Collapse
Affiliation(s)
- Shilin Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Meichen Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Xiaocui Fang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Xuechun Hu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yanlian Yang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Jie Meng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Tao Wen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jian Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Chen Wang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
10
|
IL-3 signalling in the tumour microenvironment shapes the immune response via tumour endothelial cell-derived extracellular vesicles. Pharmacol Res 2022; 179:106206. [DOI: 10.1016/j.phrs.2022.106206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/17/2022]
|
11
|
Thakral D, Gupta R, Khan A. Leukemic stem cell signatures in Acute myeloid leukemia- targeting the Guardians with novel approaches. Stem Cell Rev Rep 2022; 18:1756-1773. [DOI: 10.1007/s12015-022-10349-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2022] [Indexed: 11/09/2022]
|
12
|
Feng L, Li C, Zeng LW, Gao D, Sun YH, Zhong L, Lin H, Shu HB, Li S. MARCH3 negatively regulates IL-3-triggered inflammatory response by mediating K48-linked polyubiquitination and degradation of IL-3Rα. Signal Transduct Target Ther 2022; 7:21. [PMID: 35075102 PMCID: PMC8786845 DOI: 10.1038/s41392-021-00834-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
Interleukin-3 (IL-3) is a hematopoietic growth factor and critical regulator of inflammatory response such as sepsis. IL-3 binds to IL-3 receptor α (IL-3Rα), which is then associated with IL-3Rβ to initiate signaling. How IL-3-triggered physiological and pathological effects are regulated at the receptor level is unclear. Here, we show that the plasma membrane-associated E3 ubiquitin ligase MARCH3 negatively regulates IL-3-triggered signaling. MARCH3 is associated with IL-3Rα, mediates its K48-linked polyubiquitination at K377 and promotes its proteasomal degradation. MARCH3-deficiency promotes IL-3-triggered transcription of downstream effector genes and IL-3-induced expansion of myeloid cells. In the cecal ligation and puncture (CLP) model of sepsis, MARCH3-deficiency aggravates IL-3-ampified expression of inflammatory cytokines, organ damage and inflammatory death. Our findings suggest that regulation of IL-3Rα by MARCH3 plays an important role in IL-3-triggered physiological functions and inflammatory diseases.
Collapse
Affiliation(s)
- Lu Feng
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Chen Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Lin-Wen Zeng
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Deng Gao
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Yu-Hao Sun
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Li Zhong
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Heng Lin
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China
| | - Hong-Bing Shu
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China.
| | - Shu Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, 430071, Wuhan, China.
| |
Collapse
|
13
|
Xu S, Zhang M, Fang X, Meng J, Xing H, Yan D, Liu J, Yang Y, Wen T, Zhang W, Wang J, Wang C, Xu H. A novel CD123-targeted therapeutic peptide loaded by micellar delivery system combats refractory acute myeloid leukemia. J Hematol Oncol 2021; 14:193. [PMID: 34774070 PMCID: PMC8590286 DOI: 10.1186/s13045-021-01206-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 10/29/2021] [Indexed: 11/10/2022] Open
Abstract
Acute myeloid leukemia (AML) is a common malignant heterogeneous hematopoietic disease with very low average 5-year survival rate due to the refractory feature and high rate of relapse. CD123 is highly expressed on multiple types of AML cells, especially leukemia stem cells, and closely associated with the poor prognosis of AML. Aiming to meet the urgent demand to targeted therapeutics for the refractory AML patients, herein we synthesize a CD123 antagonistic peptide (PO-6) loaded in nanomicelles (mPO-6), and investigated its therapeutic effect and pharmacokinetics on a lab-established refractory AML mice model (AE & CKITD816V). It is shown that the PO-6 can effectively bind to the CD123+ AML cells and the micellar formulation mPO-6 increases the dissolution stability and the specific binding capacity. When injected intravenously, mPO-6 significantly prolongs the survival of the refractory AML mice by interfering CD123/IL-3 axis, evidenced by the down regulation of phosphorylation of STAT5 and PI3K/AKT and the inhibition of activated NF-κB in the nucleus, as well as by the analysis results of next generation RNA-sequencing (RNA-seq) with the bone marrow of the AML mice. The antagonistic effect leads to the significantly reduction of AML cells infiltration in the bone marrow of the AML mice. In conclusion, mPO-6 could provide a potent antagonistic therapeutic approach for targeted treatment of AML.
Collapse
Affiliation(s)
- Shilin Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Meichen Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Xiaocui Fang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing, 100190, China.,University of the Chinese Academy of Sciences, Beijing, 100190, China
| | - Jie Meng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Doudou Yan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Jian Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Yanlian Yang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing, 100190, China.,University of the Chinese Academy of Sciences, Beijing, 100190, China
| | - Tao Wen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Weiqi Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| | - Chen Wang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center of Nanoscience and Technology, Beijing, 100190, China. .,University of the Chinese Academy of Sciences, Beijing, 100190, China.
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
14
|
Abstract
The β common chain (βc) cytokine family includes granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3) and IL-5, all of which use βc as key signaling receptor subunit. GM-CSF, IL-3 and IL-5 have specific roles as hematopoietic growth factors. IL-3 binds with high affinity to the IL-3 receptor α (IL-3Rα/CD123) and then associates with the βc subunit. IL-3 is mainly synthesized by different subsets of T cells, but is also produced by several other immune [basophils, dendritic cells (DCs), mast cells, etc.] and non-immune cells (microglia and astrocytes). The IL-3Rα is also expressed by immune (basophils, eosinophils, mast cells, DCs, monocytes, and megacaryocytes) and non-immune cells (endothelial cells and neuronal cells). IL-3 is the most important growth and activating factor for human and mouse basophils, primary effector cells of allergic disorders. IL-3-activated basophils and mast cells are also involved in different chronic inflammatory disorders, infections, and several types of cancer. IL-3 induces the release of cytokines (i.e., IL-4, IL-13, CXCL8) from human basophils and preincubation of basophils with IL-3 potentiates the release of proinflammatory mediators and cytokines from IgE- and C5a-activated basophils. IL-3 synergistically potentiates IL-33-induced mediator release from human basophils. IL-3 plays a pathogenic role in several hematologic cancers and may contribute to autoimmune and cardiac disorders. Several IL-3Rα/CD123 targeting molecules have shown some efficacy in the treatment of hematologic malignancies.
Collapse
|
15
|
Bonnevaux H, Guerif S, Albrecht J, Jouannot E, De Gallier T, Beil C, Lange C, Leuschner WD, Schneider M, Lemoine C, Caron A, Amara C, Barrière C, Siavellis J, Bardet V, Luna E, Agrawal P, Drake DR, Rao E, Wonerow P, Carrez C, Blanc V, Hsu K, Wiederschain D, Fraenkel PG, Virone-Oddos A. Pre-clinical development of a novel CD3-CD123 bispecific T-cell engager using cross-over dual-variable domain (CODV) format for acute myeloid leukemia (AML) treatment. Oncoimmunology 2021; 10:1945803. [PMID: 34484869 PMCID: PMC8409758 DOI: 10.1080/2162402x.2021.1945803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Novel therapies are needed for effective treatment of AML. In the relapsed setting, prognosis is very poor despite salvage treatment with chemotherapy. Evidence suggests that leukemic stem cells (LSCs) cause relapse. The cell surface receptor CD123 is highly expressed in blast cells and LSCs from AML patients and is a potential therapeutic target. CD123 cross-over dual-variable domain T-cell engager (CD123-CODV-TCE) is a bispecific antibody with an innovative format. One arm targets the CD3εδ subunit of T-cell co-receptors on the surface of T cells, while the other targets CD123 on malignant cells, leading to cell-specific cytotoxic activity. Here, we describe the preclinical activity of CD123-CODV-TCE. CD123-CODV-TCE effectively binds to human and cynomolgus monkey CD3 and CD123 and is a highly potent T-cell engager. It mediates T-cell activation and T-cell-directed killing of AML cells in vitro. In vivo, CD123-CODV-TCE suppresses AML tumor growth in leukemia xenograft mouse models, where it achieves an effective half-life of 3.2 days, which is a significantly longer half-life compared to other bispecific antibodies with no associated Fc fragment. The in vitro safety profile is as expected for compounds with similar modes of action. These results suggest that CD123-CODV-TCE may be a promising therapy for patients with relapsed/refractory AML.
Collapse
Affiliation(s)
- Hélène Bonnevaux
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Stephane Guerif
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Jana Albrecht
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Erwan Jouannot
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Thibaud De Gallier
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Christian Beil
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Christian Lange
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Wulf Dirk Leuschner
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Marion Schneider
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Cendrine Lemoine
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Anne Caron
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Céline Amara
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Cédric Barrière
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Justine Siavellis
- Hopitaux Universitaires Paris Ile De France Ouest, Université Versailles Saint Quentin, Paris, France
| | - Valérie Bardet
- Hopitaux Universitaires Paris Ile De France Ouest, Université Versailles Saint Quentin, Paris, France
| | | | | | | | - Ercole Rao
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Peter Wonerow
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Chantal Carrez
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Véronique Blanc
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Karl Hsu
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Dmitri Wiederschain
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Paula G Fraenkel
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Angéla Virone-Oddos
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| |
Collapse
|
16
|
Agrawal V, Gbolahan OB, Stahl M, Zeidan AM, Zaid MA, Farag SS, Konig H. Vaccine and Cell-based Therapeutic Approaches in Acute Myeloid Leukemia. Curr Cancer Drug Targets 2021; 20:473-489. [PMID: 32357813 DOI: 10.2174/1568009620666200502011059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/05/2020] [Accepted: 03/29/2020] [Indexed: 12/13/2022]
Abstract
Over the past decade, our increased understanding of the interactions between the immune system and cancer cells has led to paradigm shifts in the clinical management of solid and hematologic malignancies. The incorporation of immune-targeted strategies into the treatment landscape of acute myeloid leukemia (AML), however, has been challenging. While this is in part due to the inability of the immune system to mount an effective tumor-specific immunogenic response against the heterogeneous nature of AML, the decreased immunogenicity of AML cells also represents a major obstacle in the effort to design effective immunotherapeutic strategies. In fact, AML cells have been shown to employ sophisticated escape mechanisms to evade elimination, such as direct immunosuppression of natural killer cells and decreased surface receptor expression leading to impaired recognition by the immune system. Yet, cellular and humoral immune reactions against tumor-associated antigens (TAA) of acute leukemia cells have been reported and the success of allogeneic stem cell transplantation and monoclonal antibodies in the treatment of AML clearly provides proof that an immunotherapeutic approach is feasible in the management of this disease. This review discusses the recent progress and persisting challenges in cellular immunotherapy for patients with AML.
Collapse
Affiliation(s)
- Vaibhav Agrawal
- Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Olumide B Gbolahan
- Division of Hematology and Oncology, University of Alabama School of Medicine, Birmingham, AL 35294, United States
| | - Maximilian Stahl
- Department of Medicine, Division of Hematology and Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT 06510, United States
| | - Mohammad Abu Zaid
- Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Sherif S Farag
- Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Heiko Konig
- Department of Medicine, Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|
17
|
Immunotherapy in AML: a brief review on emerging strategies. Clin Transl Oncol 2021; 23:2431-2447. [PMID: 34160771 DOI: 10.1007/s12094-021-02662-1] [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] [Received: 02/08/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022]
Abstract
Acute myeloid leukemia (AML), the most common form of leukemia amongst adults, is one of the most important hematological malignancies. Epidemiological data show both high incidence rates and low survival rates, especially in secondary cases among adults. Although classic and novel chemotherapeutic approaches have extensively improved disease prognosis and survival, the need for more personalized and target-specific methods with less side effects have been inevitable. Therefore, immunotherapeutic methods are of importance. In the following review, primarily a brief understanding of the molecular basis of the disease has been represented. Second, prior to the introduction of immunotherapeutic approaches, the entangled relationship of AML and patient's immune system has been discussed. At last, mechanistic and clinical evidence of each of the immunotherapy approaches have been covered.
Collapse
|
18
|
Aref S, Azmy E, El Ghannam D, Haroun M, Ibrahim L, Sabry M. Clinical value of CD25/CD123 co-expression in acute myeloid leukemia patients. Cancer Biomark 2021; 29:9-16. [PMID: 32417762 DOI: 10.3233/cbm-201519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND This study aimed to assess the significance of combined expression of interleukin-2 receptor (CD25) and the interleukin-3 receptor (CD123) in acute myeloid leukemia (AML) patients. METHODS The expression of CD25 and CD123 on blast cells in bone marrow samples were identified by flowcytometry in 94 patients (⩽ 60 years old) with de novo acute myeloid leukemia (AML) treated at the Mansoura University Oncology Center (MUOC). RESULTS Of the 94 samples at diagnosis there were 17 (18.1%) CD25+/CD123+ (double positive) cases; 25 (26.6%) CD25+/CD123- (single positive); 32 (34.0%) CD25-/CD123+ (single positive) cases; 20 (21.3%). CD25-/CD123- (double negative). Most of the AML patients have double CD25+/CD123+ were significantly associated with poor and intermediate risk as compared to those associated with those in the good risk group (P= 0.005). The lowest induction of remission was recorded in AML patients have double CD25+/CD123+ expression as compared to the remaining AML patient group. Study the effect of these biomarkers on the overall survival reveal that AML patients exhibited double CD25+/CD123+ expression had significantly shorter overall survival as compared to negative ones. CONCLUSION Double CD25+/CD123+ co-expression in AML patients is a dismal prognostic marker and could be used as novel biomarker for risk stratification for AML patients.
Collapse
Affiliation(s)
- Salah Aref
- Hematology Unit, Clinical Pathology Department, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | - Emaad Azmy
- Clinical Hematology Unit, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | - Doaa El Ghannam
- Hematology Unit, Clinical Pathology Department, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | - Marwa Haroun
- Hematology Unit, Clinical Pathology Department, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | - Lamiaa Ibrahim
- Hematology Unit, Clinical Pathology Department, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| | - Mohamed Sabry
- Hematology Unit, Clinical Pathology Department, Mansoura University Oncology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
19
|
Isidori A, Cerchione C, Daver N, DiNardo C, Garcia-Manero G, Konopleva M, Jabbour E, Ravandi F, Kadia T, Burguera ADLF, Romano A, Loscocco F, Visani G, Martinelli G, Kantarjian H, Curti A. Immunotherapy in Acute Myeloid Leukemia: Where We Stand. Front Oncol 2021; 11:656218. [PMID: 34041025 PMCID: PMC8143531 DOI: 10.3389/fonc.2021.656218] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
In the past few years, our improved knowledge of acute myeloid leukemia (AML) pathogenesis has led to the accelerated discovery of new drugs and the development of innovative therapeutic approaches. The role of the immune system in AML development, growth and recurrence has gained increasing interest. A better understanding of immunological escape and systemic tolerance induced by AML blasts has been achieved. The extraordinary successes of immune therapies that harness the power of T cells in solid tumors and certain hematological malignancies have provided new stimuli in this area of research. Accordingly, major efforts have been made to develop immune therapies for the treatment of AML patients. The persistence of leukemia stem cells, representing the most relevant cause of relapse, even after allogeneic stem cell transplant (allo-SCT), remains a major hurdle in the path to cure for AML patients. Several clinical trials with immune-based therapies are currently ongoing in the frontline, relapsed/refractory, post-allo-SCT and minimal residual disease/maintenance setting, with the aim to improve survival of AML patients. This review summarizes the available data with immune-based therapeutic modalities such as monoclonal antibodies (naked and conjugated), T cell engagers, adoptive T-cell therapy, adoptive-NK therapy, checkpoint blockade via PD-1/PD-L1, CTLA4, TIM3 and macrophage checkpoint blockade via the CD47/SIRPa axis, and leukemia vaccines. Combining clinical results with biological immunological findings, possibly coupled with the discovery of biomarkers predictive for response, will hopefully allow us to determine the best approaches to immunotherapy in AML.
Collapse
Affiliation(s)
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Naval Daver
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | - Courtney DiNardo
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | | | - Marina Konopleva
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | - Elias Jabbour
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | - Farhad Ravandi
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | - Tapan Kadia
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, United States
| | | | - Alessandra Romano
- Dipartimento di Chirurgia e Specialità Medico-Chirurgiche, Sezione di Ematologia, Università degli Studi di Catania, Catania, Italy
| | | | - Giuseppe Visani
- Haematology and Stem Cell Transplant Center, AORMN, Pesaro, Italy
| | - Giovanni Martinelli
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Hagop Kantarjian
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| |
Collapse
|
20
|
Li Z, Chu X, Gao L, Ling J, Xiao P, Lu J, Wang Y, He H, Li J, Hu Y, Li J, Pan J, Xiao S, Hu S. High Expression of Interleukin-3 Receptor Alpha Chain (CD123) Predicts Favorable Outcome in Pediatric B-Cell Acute Lymphoblastic Leukemia Lacking Prognosis-Defining Genomic Aberrations. Front Oncol 2021; 11:614420. [PMID: 33796456 PMCID: PMC8008053 DOI: 10.3389/fonc.2021.614420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/02/2021] [Indexed: 11/22/2022] Open
Abstract
Background Aberrant expression of CD123 (IL-3Rα) was observed in various hematological malignancies including acute lymphoblastic leukemia (ALL), which is the most common malignancy in childhood. Although widely used for minimal residual disease (MRD) monitoring, the prognostic value of CD123 has not been fully characterized in pediatric B-ALL. This retrospective study aims to evaluate the association between the CD123 expression of leukemic blasts and the outcomes of the pediatric B-ALL patients. Methods A total of 976 pediatric B-ALL, including 328 treated with CCLG-ALL-2008 protocol and 648 treated with CCCG-ALL-2015 protocol, were recruited in this retrospective study. CD123 expression was evaluated by flow cytometry. Patients with >50, 20–50, or <20% of CD123 expressing blasts were grouped into CD123high, CD123low, and CD123neg, respectively. The correlation between CD123 expression and the patients’ clinical characteristics, overall survival (OS), event-free survival (EFS), and relapse-free survival (RFS) were studied statistically. Results Of 976 pediatric B-ALL, 53.4% from the CCLG-ALL-2008 cohort and 49.2% from the CCCG-ALL-2015 cohort were CD123high. In the CCLG-ALL-2008 cohort, CD123high was significantly associated with chromosome hyperdiploidy (p < 0.0001), risk stratification (p = 0.004), and high survival rate (p = 0.005). By comparing clinical outcomes, patients with CD123high displayed favorable prognosis, with a significantly better OS (p = 0.005), EFS (p = 0.017), and RFS (p = 0.045), as compared to patients with CD123low and CD123neg. The prognostic value of CD123 expression was subsequently confirmed in the CCCG-ALL-2015 cohort. Univariate and multivariate cox regression model analysis showed that high CD123 expression was independently associated with favorable EFS (OR: 0.528; 95% CI: 0.327 to 0.853; p = 0.009) in this cohort. In patients without prognosis-defining genomic abnormalities, high CD123 expression strongly indicated superior survival rates and was identified as an independent prognosis factor for EFS and RFS in both cohorts. Conclusions A group of B-ALL lacks prognosis-defining genomic aberrations, which proposes a challenge in risk stratification. Our findings revealed that high CD123 expression of leukemic blasts was associated with favorable clinical outcomes in pediatric B-ALL and CD123 could serve as a promising prognosis predictor, especially in patients without prognosis-defining genetic aberrations.
Collapse
Affiliation(s)
- Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Xinran Chu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Li Gao
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jing Ling
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Peifang Xiao
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jun Lu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Yi Wang
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Hailong He
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jianqin Li
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Yixin Hu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jie Li
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Shaoyan Hu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
21
|
Chimeric antigen receptor T cell therapies for acute myeloid leukemia. Front Med 2020; 14:701-710. [PMID: 33263835 DOI: 10.1007/s11684-020-0763-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/16/2020] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor T cell (CAR T) therapies have achieved unprecedented efficacy in B-cell tumors, prompting scientists and doctors to exploit this strategy to treat other tumor types. Acute myeloid leukemia (AML) is a group of heterogeneous myeloid malignancies. Relapse remains the main cause of treatment failure, especially for patients with intermediate or high risk stratification. Allogeneic hematopoietic stem cell transplantation could be an effective therapy because of the graft-versus-leukemia effect, which unfortunately puts the patient at risk of serious complications, such as graft-versus-host disease. Although the identification of an ideal target antigen for AML is challenging, CAR T therapy remains a highly promising strategy for AML patients, particularly for those who are ineligible to receive a transplantation or have positive minimal residual disease. In this review, we focus on the most recent and promising advances in CAR T therapies for AML.
Collapse
|
22
|
Dong X, Zhang D, Zhang J, Chen X, Zhang Y, Zhang Y, Zhou X, Chen T, Zhou H. Immune prognostic risk score model in acute myeloid leukemia with normal karyotype. Oncol Lett 2020; 20:380. [PMID: 33154778 DOI: 10.3892/ol.2020.12243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/08/2020] [Indexed: 01/14/2023] Open
Abstract
Acute myeloid leukemia with normal karyotype (NK-AML) is a group of diseases with high heterogeneity and immunological processes are significantly associated with its initiation and development. The implication of the immunogenomic landscape in the prognosis of patients with NK-AML has remained largely elusive. In the present study, the expression profiles of immune-related genes (IRGs) were examined and their association with overall survival (OS) was determined in 60 patients with NK-AML from The Cancer Genome Atlas dataset and 104 patients from the Gene Expression Omnibus (GEO) dataset no. GSE71014. Univariate Cox regression analysis was used to identify 42 and 203 IRGs in the two respective cohorts, which were significantly associated with OS in NK-AML. A risk model was constructed based on the regression coefficient and expression values of nine survival-associated IRGs shared between the two datasets [zinc finger CCCH-type containing, antiviral 1 like; transferrin receptor; suppressor of cytokine signaling 1; ELAV like RNA binding protein 1; roundabout guidance receptor 3; unc-93 homolog B1, Toll-like receptor signaling regulator; protein tyrosine phosphatase non-receptor type 6; interleukin 2 receptor subunit alpha (IL2RA) and IL3RA]. Using this risk model, patients with NK-AML may be divided into high- and low-risk groups in prognostic predictions. The area under the receiver operating characteristic curve for predicting OS was 0.793. The prognostic role of this risk model was successfully verified in another independent cohort (GEO dataset no. GSE71014). The prognostic risk score was positively associated with age and fms related receptor tyrosine kinase 3 mutation and correlated with infiltration by T regulatory cells. In conclusion, the results of the present study provided an IRG score model for prognostic stratification of adult patients with NK-AML, as well as further insight into the implication of IRGs in NK-AML that may lead to the development of novel immunotherapy approaches for this disease.
Collapse
Affiliation(s)
- Xiaomin Dong
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| | - Danyang Zhang
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| | - Juan Zhang
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| | - Xiaolei Chen
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| | - Yue Zhang
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| | - Yong Zhang
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| | - Xiaohuan Zhou
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| | - Tingting Chen
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| | - Hebing Zhou
- Department of Hematology, The Affiliated Beijing Luhe Hospital of Capital Medical University, Beijing 101149, P.R. China
| |
Collapse
|
23
|
Cangini D, Silimbani P, Cafaro A, Giannini MB, Masini C, Ghelli Luserna Di Rorà A, Simonetti G, Martinelli G, Cerchione C. Tagraxofusp and anti-CD123 in blastic plasmacytoid dendritic cell neoplasm: a new hope. Minerva Med 2020; 111:467-477. [PMID: 32955827 DOI: 10.23736/s0026-4806.20.07018-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematological malignancy, characterized by poor prognosis if treated with conventional therapy. Allogenic hematologic stem cell transplant can improve survival and can be curative, but it is available in a small percentage of patients given that the median age at diagnosis is 70 years. In this scenario it is assumed that only the development of precision medicine-driven therapy will change BPDCN patient prognosis. CD123 (the α-subunit of interleukin (IL)-3 receptor) is over-expressed on BPDCN cells surface and seems to be the ideal marker to develop antibody-based therapies. Tagraxofusp (Elzonris<sup>®</sup>), a recombinant immunotoxin consisting of human interleukin-3 fused to a truncated diphtheria toxin, has been approved by FDA in December 2018 for the treatment of BPDCN in adult and pediatric patients. tagraxofusp has shown promising clinical activity, with a high overall response rate and quite manageable safety profile even in elderly patients. It seems to improve overall survival too, but comparative trials are necessary to confirm this. Adverse events are commonly reported and the most important are transaminitis, thrombocytopenia and capillary leak syndrome (CLS). Therefore, to prevent the onset of severe CLS is recommended to reserve tagraxofusp for patients with preserved hepatic and cardiac functions, and to strictly observe serum albumin level. Further studies are required to resolve many several unanswered questions about tagraxofusp. In this review, we will resume and discuss pharmacological characteristic of tagraxofusp, results of clinical trials leading to its approval by FDA in 2018 and future perspectives about its use in BPDCN and other hematological malignancies.
Collapse
Affiliation(s)
- Delia Cangini
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Paolo Silimbani
- Unit of Oncological Pharmacy, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Alessandro Cafaro
- Unit of Oncological Pharmacy, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Maria B Giannini
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Carla Masini
- Unit of Oncological Pharmacy, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Andrea Ghelli Luserna Di Rorà
- Laboratory of Biosciences, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Giorgia Simonetti
- Laboratory of Biosciences, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Claudio Cerchione
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy -
| |
Collapse
|
24
|
Schulte-Schrepping J, Reusch N, Paclik D, Baßler K, Schlickeiser S, Zhang B, Krämer B, Krammer T, Brumhard S, Bonaguro L, De Domenico E, Wendisch D, Grasshoff M, Kapellos TS, Beckstette M, Pecht T, Saglam A, Dietrich O, Mei HE, Schulz AR, Conrad C, Kunkel D, Vafadarnejad E, Xu CJ, Horne A, Herbert M, Drews A, Thibeault C, Pfeiffer M, Hippenstiel S, Hocke A, Müller-Redetzky H, Heim KM, Machleidt F, Uhrig A, Bosquillon de Jarcy L, Jürgens L, Stegemann M, Glösenkamp CR, Volk HD, Goffinet C, Landthaler M, Wyler E, Georg P, Schneider M, Dang-Heine C, Neuwinger N, Kappert K, Tauber R, Corman V, Raabe J, Kaiser KM, Vinh MT, Rieke G, Meisel C, Ulas T, Becker M, Geffers R, Witzenrath M, Drosten C, Suttorp N, von Kalle C, Kurth F, Händler K, Schultze JL, Aschenbrenner AC, Li Y, Nattermann J, Sawitzki B, Saliba AE, Sander LE. Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment. Cell 2020; 182:1419-1440.e23. [PMID: 32810438 PMCID: PMC7405822 DOI: 10.1016/j.cell.2020.08.001] [Citation(s) in RCA: 1032] [Impact Index Per Article: 206.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/13/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a mild to moderate respiratory tract infection, however, a subset of patients progress to severe disease and respiratory failure. The mechanism of protective immunity in mild forms and the pathogenesis of severe COVID-19 associated with increased neutrophil counts and dysregulated immune responses remain unclear. In a dual-center, two-cohort study, we combined single-cell RNA-sequencing and single-cell proteomics of whole-blood and peripheral-blood mononuclear cells to determine changes in immune cell composition and activation in mild versus severe COVID-19 (242 samples from 109 individuals) over time. HLA-DRhiCD11chi inflammatory monocytes with an interferon-stimulated gene signature were elevated in mild COVID-19. Severe COVID-19 was marked by occurrence of neutrophil precursors, as evidence of emergency myelopoiesis, dysfunctional mature neutrophils, and HLA-DRlo monocytes. Our study provides detailed insights into the systemic immune response to SARS-CoV-2 infection and reveals profound alterations in the myeloid cell compartment associated with severe COVID-19.
Collapse
Affiliation(s)
| | - Nico Reusch
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Daniela Paclik
- Institute of Medical Immunology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Kevin Baßler
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Stephan Schlickeiser
- Institute of Medical Immunology, Charité, Universitätsmedizin Berlin, Berlin, Germany; BIH Center for Regenerative Therapies, Charité, Universitätsmedizin Berlin, and Berlin Institute of Health (BIH) Berlin, Germany
| | - Bowen Zhang
- Centre for Individualised Infection Medicine (CiiM) and TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Benjamin Krämer
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Tobias Krammer
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Sophia Brumhard
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Lorenzo Bonaguro
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Elena De Domenico
- German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, and University of Bonn, Bonn, Germany
| | - Daniel Wendisch
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Grasshoff
- Centre for Individualised Infection Medicine (CiiM) and TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | | | - Michael Beckstette
- Centre for Individualised Infection Medicine (CiiM) and TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Tal Pecht
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Adem Saglam
- German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, and University of Bonn, Bonn, Germany
| | - Oliver Dietrich
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Henrik E Mei
- Mass Cytometry Lab, DRFZ Berlin, a Leibniz Institute, Berlin, Germany
| | - Axel R Schulz
- Mass Cytometry Lab, DRFZ Berlin, a Leibniz Institute, Berlin, Germany
| | - Claudia Conrad
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Désirée Kunkel
- Flow and Mass Cytometry Core Facility, Charité, Universitätsmedizin Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Ehsan Vafadarnejad
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM) and TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Arik Horne
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Miriam Herbert
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Anna Drews
- German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, and University of Bonn, Bonn, Germany
| | - Charlotte Thibeault
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Pfeiffer
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL)
| | - Andreas Hocke
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL)
| | - Holger Müller-Redetzky
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Katrin-Moira Heim
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Machleidt
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Uhrig
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Laure Bosquillon de Jarcy
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Linda Jürgens
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Miriam Stegemann
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph R Glösenkamp
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Dieter Volk
- Institute of Medical Immunology, Charité, Universitätsmedizin Berlin, Berlin, Germany; BIH Center for Regenerative Therapies, Charité, Universitätsmedizin Berlin, and Berlin Institute of Health (BIH) Berlin, Germany; Department of Immunology, Labor Berlin-Charité Vivantes, Berlin, Germany
| | - Christine Goffinet
- Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Philipp Georg
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Schneider
- Institute of Medical Immunology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Chantip Dang-Heine
- Clinical Study Center (CSC), Charité, Universitätsmedizin Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nick Neuwinger
- Department of Immunology, Labor Berlin-Charité Vivantes, Berlin, Germany; Institute of Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Kai Kappert
- Department of Immunology, Labor Berlin-Charité Vivantes, Berlin, Germany; Institute of Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Rudolf Tauber
- Department of Immunology, Labor Berlin-Charité Vivantes, Berlin, Germany; Institute of Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Victor Corman
- Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jan Raabe
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Kim Melanie Kaiser
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Michael To Vinh
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Gereon Rieke
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Christian Meisel
- Institute of Medical Immunology, Charité, Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology, Labor Berlin-Charité Vivantes, Berlin, Germany
| | - Thomas Ulas
- German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, and University of Bonn, Bonn, Germany
| | - Matthias Becker
- German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, and University of Bonn, Bonn, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz-Center for Infection Research (HZI), Braunschweig, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL)
| | - Christian Drosten
- Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany; German Center for Infection Research (DZIF)
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL)
| | - Christof von Kalle
- Clinical Study Center (CSC), Charité, Universitätsmedizin Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany; I. Department of Medicine, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Kristian Händler
- German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), PRECISE Platform for Genomics and Epigenomics at DZNE, and University of Bonn, Bonn, Germany.
| | - Anna C Aschenbrenner
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM) and TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany; German Center for Infection Research (DZIF)
| | - Birgit Sawitzki
- Institute of Medical Immunology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Leif Erik Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL)
| |
Collapse
|
25
|
AML risk stratification models utilizing ELN-2017 guidelines and additional prognostic factors: a SWOG report. Biomark Res 2020; 8:29. [PMID: 32817791 PMCID: PMC7425159 DOI: 10.1186/s40364-020-00208-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/27/2020] [Indexed: 01/01/2023] Open
Abstract
Background The recently updated European LeukemiaNet risk stratification guidelines combine cytogenetic abnormalities and genetic mutations to provide the means to triage patients with acute myeloid leukemia for optimal therapies. Despite the identification of many prognostic factors, relatively few have made their way into clinical practice. Methods In order to assess and improve the performance of the European LeukemiaNet guidelines, we developed novel prognostic models using the biomarkers from the guidelines, age, performance status and select transcript biomarkers. The models were developed separately for mononuclear cells and viable leukemic blasts from previously untreated acute myeloid leukemia patients (discovery cohort, N = 185) who received intensive chemotherapy. Models were validated in an independent set of similarly treated patients (validation cohort, N = 166). Results Models using European LeukemiaNet guidelines were significantly associated with clinical outcomes and, therefore, utilized as a baseline for comparisons. Models incorporating age and expression of select transcripts with biomarkers from European LeukemiaNet guidelines demonstrated higher area under the curve and C-statistics but did not show a substantial improvement in performance in the validation cohort. Subset analyses demonstrated that models using only the European LeukemiaNet guidelines were a better fit for younger patients (age < 55) than for older patients. Models integrating age and European LeukemiaNet guidelines visually showed more separation between risk groups in older patients. Models excluding results for ASXL1, CEBPA, RUNX1 and TP53, demonstrated that these mutations provide a limited overall contribution to risk stratification across the entire population, given the low frequency of mutations and confounding risk factors. Conclusions While European LeukemiaNet guidelines remain a critical tool for triaging patients with acute myeloid leukemia, the findings illustrate the need for additional prognostic factors, including age, to improve risk stratification.
Collapse
|
26
|
Aldoss I, Clark M, Song JY, Pullarkat V. Targeting the alpha subunit of IL-3 receptor (CD123) in patients with acute leukemia. Hum Vaccin Immunother 2020; 16:2341-2348. [PMID: 32692611 DOI: 10.1080/21645515.2020.1788299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The IL-3 alpha chain receptor (CD123) is a cell surface protein that is widely expressed by various subtypes of acute leukemia, including acute myeloid leukemia (AML), acute lymphoblastic leukemia and blastic plasmacytoid dendritic cell neoplasm. Notably, CD123 is preferentially overexpressed in leukemia stem cells (LSC) in contrast to normal hematopoietic stem cells, and this differential expression allows for the selective eradication of LSC and leukemic blasts through therapeutic targeting of CD123, with less impact on hematopoietic cells. The level of CD123 expression in AML correlates with both treatment response and outcomes. Therefore, targeting CD123 represents a promising universal therapeutic target in advanced acute leukemias irrespective of the individual leukemia phenotype. There are currently 31 ongoing clinical trials examining the utility of CD123-based targeted therapies. Here we focus our review on current efforts to target CD123 in acute leukemia through various therapeutic constructs.
Collapse
Affiliation(s)
- Ibrahim Aldoss
- Gehr Family Center for Leukemia Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center , Duarte, CA, USA
| | - Mary Clark
- Department of Clinical and Translational Project Development, City of Hope National Medical Center , Duarte, CA, USA
| | - Joo Y Song
- Department of Pathology, City of Hope National Medical Center , Duarte, CA, USA
| | - Vinod Pullarkat
- Gehr Family Center for Leukemia Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center , Duarte, CA, USA
| |
Collapse
|
27
|
Baroni ML, Sanchez Martinez D, Gutierrez Aguera F, Roca Ho H, Castella M, Zanetti SR, Velasco Hernandez T, Diaz de la Guardia R, Castaño J, Anguita E, Vives S, Nomdedeu J, Lapillone H, Bras AE, van der Velden VHJ, Junca J, Marin P, Bataller A, Esteve J, Vick B, Jeremias I, Lopez A, Sorigue M, Bueno C, Menendez P. 41BB-based and CD28-based CD123-redirected T-cells ablate human normal hematopoiesis in vivo. J Immunother Cancer 2020; 8:e000845. [PMID: 32527933 PMCID: PMC7292050 DOI: 10.1136/jitc-2020-000845] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematopoietic malignancy which is biologically, phenotypically and genetically very heterogeneous. Outcome of patients with AML remains dismal, highlighting the need for improved, less toxic therapies. Chimeric antigen receptor T-cell (CART) immunotherapies for patients with refractory or relapse (R/R) AML are challenging because of the absence of a universal pan-AML target antigen and the shared expression of target antigens with normal hematopoietic stem/progenitor cells (HSPCs), which may lead to life-threating on-target/off-tumor cytotoxicity. CD33-redirected and CD123-redirected CARTs for AML are in advanced preclinical and clinical development, and they exhibit robust antileukemic activity. However, preclinical and clinical controversy exists on whether such CARTs are myeloablative. METHODS We set out to comparatively characterize in vitro and in vivo the efficacy and safety of 41BB-based and CD28-based CARCD123. We analyzed 97 diagnostic and relapse AML primary samples to investigate whether CD123 is a suitable immunotherapeutic target, and we used several xenograft models and in vitro assays to assess the myeloablative potential of our second-generation CD123 CARTs. RESULTS Here, we show that CD123 represents a bona fide target for AML and show that both 41BB-based and CD28-based CD123 CARTs are very efficient in eliminating both AML cell lines and primary cells in vitro and in vivo. However, both 41BB-based and CD28-based CD123 CARTs ablate normal human hematopoiesis and prevent the establishment of de novo hematopoietic reconstitution by targeting both immature and myeloid HSPCs. CONCLUSIONS This study calls for caution when clinically implementing CD123 CARTs, encouraging its preferential use as a bridge to allo-HSCT in patients with R/R AML.
Collapse
Affiliation(s)
- Matteo Libero Baroni
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Diego Sanchez Martinez
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | | | - Heleia Roca Ho
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Maria Castella
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Samanta Romina Zanetti
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Talia Velasco Hernandez
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | | | - Julio Castaño
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Eduardo Anguita
- Hematology and Hemotherapy Department, Hospital Clinico Universitario San Carlos Instituto Cardiovascular, Madrid, Comunidad de Madrid, Spain
| | - Susana Vives
- Hematology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Catalunya, Spain
| | - Josep Nomdedeu
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
| | - Helene Lapillone
- Centre de Recherce Saint-Antoine, Armand-Trousseau Childrens Hospital, Paris, Île-de-France, France
| | - Anne E Bras
- Immunology Department, Erasmus Medical Center, Rotterdam, Zuid-Holland, Netherlands
| | | | - Jordi Junca
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
- Hematology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Catalunya, Spain
| | - Pedro Marin
- Hematology Department, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Alex Bataller
- Hematology Department, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Jordi Esteve
- Hematology Department, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Binje Vick
- Helmholtz Center, Munich German Research Center for Environmental Health, Neuherberg, Bayern, Germany
| | - Irmela Jeremias
- Helmholtz Center, Munich German Research Center for Environmental Health, Neuherberg, Bayern, Germany
- Pediatrics Department, Munich University Hospital Dr von Hauner Children's Hospital, Munchen, Bayern, Germany
| | - Angel Lopez
- Human Immunology Department, Centre for Cancer Biology, Adelaide, South Australia, Australia
| | - Marc Sorigue
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
- Hematology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Catalunya, Spain
| | - Clara Bueno
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
- Centro de investigación en Red-Oncología, CIBERONC, Comunidad de Madrid, Madrid, Spain
| | - Pablo Menendez
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
- Centro de investigación en Red-Oncología, CIBERONC, Comunidad de Madrid, Madrid, Spain
- Instituciò Catalana de Recerca i Estudis Avançats, ICREA, Barcelona, Catalunya, Spain
| |
Collapse
|
28
|
Hammond D, Pemmaraju N. Tagraxofusp for Blastic Plasmacytoid Dendritic Cell Neoplasm. Hematol Oncol Clin North Am 2020; 34:565-574. [PMID: 32336420 DOI: 10.1016/j.hoc.2020.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tagraxofusp, a CD123-targeted immunotoxin, is the first FDA-approved treatment for patients 2 years and older with blastic plasmacytoid dendritic cell neoplasm (BPDCN). It has been shown to be safe and effective in treatment-naïve and previously treated adult patients, with high rates of successful bridging to hematopoietic stem cell transplantation. The pediatric experience is more limited but demonstrates safety. Given the risk of potentially fatal capillary leak syndrome with tagraxofusp, judicious patient selection is recommended. Combination therapy with hypomethylating agents and/or BCL-2 inhibitors are rational next lines of investigation, especially in patients ineligible to receive high-dose chemotherapy.
Collapse
Affiliation(s)
- Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA. https://twitter.com/DanielleHammo20
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
29
|
Alfayez M, Konopleva M, Pemmaraju N. Role of tagraxofusp in treating blastic plasmacytoid dendritic cell neoplasm (BPDCN). Expert Opin Biol Ther 2019; 20:115-123. [PMID: 31801379 DOI: 10.1080/14712598.2020.1701651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Advances and drug development in rare diseases, such as blastic plasmacytoid dendritic cell neoplasm (BPDCN), has historically been limited by small numbers of patients in the target population. In recent years, the development of tagraxofusp (SL-401) (ELZONRIS, Stemline Therapeutics) for the treatment of adult and pediatric BPDCN has been a successful story that led to US FDA approval in December 2018.Areas covered: In this evaluation of tagraxofusp, we briefly review chemistry; pharmacokinetics and pharmacodynamics, as we focus on the clinical experience and future directions.Expert Opinion: Tagraxofusp has been a welcome new addition and a successful initial development step in the targeted treatment of BPDCN. In phase I/II clinical trial, major responses were observed in 90% of treatment-naïve patients, with 72% of the responses observed as complete remissions. Limitations on the usage of tagraxofusp and strategies to handle those limitations were further explored in this review.
Collapse
Affiliation(s)
- Mansour Alfayez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
30
|
Shang Y, Zhou F. Current Advances in Immunotherapy for Acute Leukemia: An Overview of Antibody, Chimeric Antigen Receptor, Immune Checkpoint, and Natural Killer. Front Oncol 2019; 9:917. [PMID: 31616632 PMCID: PMC6763689 DOI: 10.3389/fonc.2019.00917] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022] Open
Abstract
Recently, due to the application of hematopoietic stem cell transplantation and small molecule inhibitor, the survival of acute leukemia is prolonged. However, the 5 year survival rate remains low due to a high incidence of relapse. Immunotherapy is expected to improve the prognosis of patients with relapsed or refractory hematological malignancies because it does not rely on the cytotoxic mechanisms of conventional therapy. In this paper, the advances of immunotherapy in acute leukemia are reviewed from the aspects of Antibody including Unconjugated antibodies, Antibody-drug conjugate and Bispecific antibody, Chimeric Antigen Receptor (CARs), Immune checkpoint, Natural killer cells. The immunological features, mechanisms and limitation in clinic will be described.
Collapse
Affiliation(s)
- Yufeng Shang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
31
|
CD123 as a Therapeutic Target in the Treatment of Hematological Malignancies. Cancers (Basel) 2019; 11:cancers11091358. [PMID: 31547472 PMCID: PMC6769702 DOI: 10.3390/cancers11091358] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
The interleukin-3 receptor alpha chain (IL-3Rα), more commonly referred to as CD123, is widely overexpressed in various hematological malignancies, including acute myeloid leukemia (AML), B-cell acute lymphoblastic leukemia, hairy cell leukemia, Hodgkin lymphoma and particularly, blastic plasmacytoid dendritic neoplasm (BPDCN). Importantly, CD123 is expressed at both the level of leukemic stem cells (LSCs) and more differentiated leukemic blasts, which makes CD123 an attractive therapeutic target. Various agents have been developed as drugs able to target CD123 on malignant leukemic cells and on the normal counterpart. Tagraxofusp (SL401, Stemline Therapeutics), a recombinant protein composed of a truncated diphtheria toxin payload fused to IL-3, was approved for use in patients with BPDCN in December of 2018 and showed some clinical activity in AML. Different monoclonal antibodies directed against CD123 are under evaluation as antileukemic drugs, showing promising results either for the treatment of AML minimal residual disease or of relapsing/refractory AML or BPDCN. Finally, recent studies are exploring T cell expressing CD123 chimeric antigen receptor-modified T-cells (CAR T) as a new immunotherapy for the treatment of refractory/relapsing AML and BPDCN. In December of 2018, MB-102 CD123 CAR T developed by Mustang Bio Inc. received the Orphan Drug Designation for the treatment of BPDCN. In conclusion, these recent studies strongly support CD123 as an important therapeutic target for the treatment of BPDCN, while a possible in the treatment of AML and other hematological malignancies will have to be evaluated by in the ongoing clinical studies.
Collapse
|
32
|
Molecular mechanisms for stemness maintenance of acute myeloid leukemia stem cells. BLOOD SCIENCE 2019; 1:77-83. [PMID: 35402786 PMCID: PMC8975089 DOI: 10.1097/bs9.0000000000000020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/17/2019] [Indexed: 11/26/2022] Open
Abstract
Human acute myeloid leukemia (AML) is a fatal hematologic malignancy characterized with accumulation of myeloid blasts and differentiation arrest. The development of AML is associated with a serial of genetic and epigenetic alterations mainly occurred in hematopoietic stem and progenitor cells (HSPCs), which change HSPC state at the molecular and cellular levels and transform them into leukemia stem cells (LSCs). LSCs play critical roles in leukemia initiation, progression, and relapse, and need to be eradicated to achieve a cure in clinic. Key to successfully targeting LSCs is to fully understand the unique cellular and molecular mechanisms for maintaining their stemness. Here, we discuss LSCs in AML with a focus on identification of unique biological features of these stem cells to decipher the molecular mechanisms of LSC maintenance.
Collapse
|
33
|
Arai N, Homma M, Abe M, Baba Y, Murai S, Watanuki M, Kawaguchi Y, Fujiwara S, Kabasawa N, Tsukamoto H, Uto Y, Ariizumi H, Yanagisawa K, Hattori N, Saito B, Shiozawa E, Harada H, Yamochi-Onizuka T, Nakamaki T, Takimoto M. Impact of CD123 expression, analyzed by immunohistochemistry, on clinical outcomes in patients with acute myeloid leukemia. Int J Hematol 2019; 109:539-544. [PMID: 30847774 DOI: 10.1007/s12185-019-02616-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 12/16/2022]
Abstract
Aberrant expression of the interleukin-3 receptor alpha chain (IL3RA or CD123) is frequently observed in patients with a subset of leukemic disorders, including acute myeloid leukemia (AML), particularly in leukemia stem cells. We analyzed the relationships between immunohistochemical (IHC) expression, including that of CD123, and clinical outcomes. This study involved a retrospective analysis of 48 patients diagnosed with de novo AML (M0-M5, n = 48) at our hospital between February 2008 and September 2015. Among patients with de novo AML, CD123 expression was associated with a failure to achieve complete response (CR) to initial induction chemotherapy (P = 0.044) and poor overall survival (OS) (P = 0.036). This is the first study using IHC to demonstrate that CD123 expression is associated with a poor CR rate and poor OS in de novo AML patients. These results support previous reports using flow cytometry (FCM). CD123 expression may thus be useful for assessing AML patients' prognoses. At the time of diagnosis, CD123 expression analysis using IHC may represent a clinically useful assessment for de novo AML patients.
Collapse
Affiliation(s)
- Nana Arai
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan. .,Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan.
| | - Mayumi Homma
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| | - Maasa Abe
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Yuta Baba
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - So Murai
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Megumi Watanuki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Yukiko Kawaguchi
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Shun Fujiwara
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Nobuyuki Kabasawa
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Hiroyuki Tsukamoto
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Yui Uto
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Hirotsugu Ariizumi
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Kouji Yanagisawa
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Norimichi Hattori
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Bungo Saito
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Eisuke Shiozawa
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| | - Hiroshi Harada
- Division of Hematology, Department of Medicine, Showa University Fujigaoka Hospital, Kanagawa, Japan
| | | | - Tsuyoshi Nakamaki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Masafumi Takimoto
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
Alkharabsheh O, Frankel AE. Clinical Activity and Tolerability of SL-401 (Tagraxofusp): Recombinant Diphtheria Toxin and Interleukin-3 in Hematologic Malignancies. Biomedicines 2019; 7:biomedicines7010006. [PMID: 30621282 PMCID: PMC6466259 DOI: 10.3390/biomedicines7010006] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/26/2018] [Accepted: 01/03/2019] [Indexed: 12/24/2022] Open
Abstract
Overcoming the leukemia stem cell resistance to intensive chemotherapy has been an area of extensive research over the last two decades. Advances and greater understanding of the molecular biology of leukemia stem cells are in rapid progress. Targeted therapies are currently being used in clinical practice with reasonable response rates, but a cure is being achieved in only a small percentage of patients, most likely due to tumor mutational heterogeneity. A genetically engineered diphtheria toxin fused with interleukin-3 (SL-401 or tagraxofusp) has shown robust activity in blastic plasmacytoid dendritic cell neoplasm and promising response rates in different myeloid malignancies, including eradication of minimal residual disease. Multiple clinical trials are being conducted using this drug and the preliminary results are encouraging. This article reviews the clinical trials for SL-401, its mechanism of action, clinical activity, and the adverse event profile.
Collapse
Affiliation(s)
- Omar Alkharabsheh
- Division of Medical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| | - Arthur E Frankel
- Division of Medical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA.
| |
Collapse
|
35
|
Abstract
Transposon mutagenesis has emerged as a powerful methodology for functionally annotating cancer genomes. Although in vivo transposon-mediated forward genetic screens have proven to be valuable for cancer gene identification, they are also time consuming and resource intensive. To facilitate the rapid and cost-effective identification of genes that regulate tumor-promoting pathways, we developed a complementary ex vivo transposon mutagenesis approach wherein human or mouse cells growing in culture are mutagenized and screened for the acquisition of specific phenotypes in vitro or in vivo, such as growth factor independence or tumor-forming ability. This approach allows discovery of both gain- and loss-of-function mutations in the same screen. Transposon insertions sites are recovered by high-throughput sequencing. We recently applied this system to comprehensively identify and validate genes that promote growth factor independence and transformation of murine Ba/F3 cells. Here we describe a method for performing ex vivo Sleeping Beauty-mediated mutagenesis screens in these cells, which may be adapted for the acquisition of many different phenotypes in distinct cell types.
Collapse
|
36
|
Lee HJ, Jedrychowski MP, Vinayagam A, Wu N, Shyh-Chang N, Hu Y, Min-Wen C, Moore JK, Asara JM, Lyssiotis CA, Perrimon N, Gygi SP, Cantley LC, Kirschner MW. Proteomic and Metabolomic Characterization of a Mammalian Cellular Transition from Quiescence to Proliferation. Cell Rep 2018; 20:721-736. [PMID: 28723573 DOI: 10.1016/j.celrep.2017.06.074] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/22/2017] [Accepted: 06/25/2017] [Indexed: 12/28/2022] Open
Abstract
There exist similarities and differences in metabolism and physiology between normal proliferative cells and tumor cells. Once a cell enters the cell cycle, metabolic machinery is engaged to facilitate various processes. The kinetics and regulation of these metabolic changes have not been properly evaluated. To correlate the orchestration of these processes with the cell cycle, we analyzed the transition from quiescence to proliferation of a non-malignant murine pro-B lymphocyte cell line in response to IL-3. Using multiplex mass-spectrometry-based proteomics, we show that the transition to proliferation shares features generally attributed to cancer cells: upregulation of glycolysis, lipid metabolism, amino-acid synthesis, and nucleotide synthesis and downregulation of oxidative phosphorylation and the urea cycle. Furthermore, metabolomic profiling of this transition reveals similarities to cancer-related metabolic pathways. In particular, we find that methionine is consumed at a higher rate than that of other essential amino acids, with a potential link to maintenance of the epigenome.
Collapse
Affiliation(s)
- Ho-Joon Lee
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Ning Wu
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Ng Shyh-Chang
- Stem Cell & Regenerative Biology, Genome Institute of Singapore, S138672 Singapore, Singapore
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Chua Min-Wen
- Stem Cell & Regenerative Biology, Genome Institute of Singapore, S138672 Singapore, Singapore
| | - Jodene K Moore
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - John M Asara
- Division of Signal Transduction, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Costas A Lyssiotis
- Division of Gastroenterology, Department of Molecular and Integrative Physiology and Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Marc W Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Przespolewski A, Szeles A, Wang ES. Advances in immunotherapy for acute myeloid leukemia. Future Oncol 2018. [DOI: 10.2217/fon-2017-0459] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Evasion of the host immune system is a key mechanism to promote malignant progression. Therapeutically targeting immune pathways has radically changed the treatment paradigm for solid and lymphoid tumors but has yet to be approved for myeloid malignancies. Here, we summarize the most recent advances in immunotherapy for acute myeloid leukemia. Topics reviewed here include adoptive cellular approaches (chimeric antigen receptor-T cells, natural killer and other immune cells), checkpoint inhibitors (anti-PD-1/PD-L1, anti-CTLA-4 and TIM-3) and vaccines (WT-1, HLA-A2 and hTERT). Emphasis is placed on agents with clear evidence of tumor-specific immune responses and/or clinical activity in early-phase trials. Despite concerns regarding heterogeneous antigen expression and cytokine release syndrome, immunotherapy remains a highly promising strategy for acute myeloid leukemia, particularly transplant-ineligible patients and minimal residual disease states.
Collapse
Affiliation(s)
- Amanda Przespolewski
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Andras Szeles
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Semmelweis University, Budapest, Üllői út 26, 1085, Hungary
| | - Eunice S Wang
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
38
|
Weng S, Stoner SA, Zhang DE. Sex chromosome loss and the pseudoautosomal region genes in hematological malignancies. Oncotarget 2018; 7:72356-72372. [PMID: 27655702 PMCID: PMC5342167 DOI: 10.18632/oncotarget.12050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/07/2016] [Indexed: 11/25/2022] Open
Abstract
Cytogenetic aberrations, such as chromosomal translocations, aneuploidy, and amplifications, are frequently detected in hematological malignancies. For many of the common autosomal aberrations, the mechanisms underlying their roles in cancer development have been well-characterized. On the contrary, although loss of a sex chromosome is observed in a broad range of hematological malignancies, how it cooperates in disease development is less understood. Nevertheless, it has been postulated that tumor suppressor genes reside on the sex chromosomes. Although the X and Y sex chromosomes are highly divergent, the pseudoautosomal regions are homologous between both chromosomes. Here, we review what is currently known about the pseudoautosomal region genes in the hematological system. Additionally, we discuss implications for haploinsufficiency of critical pseudoautosomal region sex chromosome genes, driven by sex chromosome loss, in promoting hematological malignancies. Because mechanistic studies on disease development rely heavily on murine models, we also discuss the challenges and caveats of existing models, and propose alternatives for examining the involvement of pseudoautosomal region genes and loss of a sex chromosome in vivo. With the widespread detection of loss of a sex chromosome in different hematological malignances, the elucidation of the role of pseudoautosomal region genes in the development and progression of these diseases would be invaluable to the field.
Collapse
Affiliation(s)
- Stephanie Weng
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Samuel A Stoner
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Dong-Er Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.,Department of Pathology and Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
39
|
Shahrabi S, Khodadi E, Saba F, Shahjahani M, Saki N. Sex chromosome changes in leukemia: cytogenetics and molecular aspects. Hematology 2017; 23:139-147. [DOI: 10.1080/10245332.2017.1375063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Elahe Khodadi
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fakhredin Saba
- Department of Laboratory Science, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Shahjahani
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
40
|
High CD123 levels enhance proliferation in response to IL-3, but reduce chemotaxis by downregulating CXCR4 expression. Blood Adv 2017; 1:1067-1079. [PMID: 29296749 DOI: 10.1182/bloodadvances.2016002931] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 05/21/2017] [Indexed: 11/20/2022] Open
Abstract
High expression of the α chain of the interleukin-3 receptor (IL-3Rα; CD123) is a hallmark of acute myeloid leukemia (AML) leukemic stem cells (LSCs). Elevated CD123 expression is part of the diagnostic immunophenotyping of myeloid leukemia, and higher expression is associated with poor prognosis. However, the biological basis of the poorer prognosis is unclear, and may include heightened IL-3 signaling and non-cell autonomous interactions with the bone marrow (BM) microenvironment. We used TF-1 cells expressing different levels of CD123 and found elevated CD123 levels amplified the proliferative response to exogenous IL-3 and maintained viability in reducing IL-3 concentrations. This was associated with stronger activation of STAT5, Akt, and extracellular signal-regulated kinase 1/2 in vitro. Surprisingly, in vivo e14.5 fetal liver cells transduced with retroviral constructs to express high CD123 failed to engraft in syngeneic recipients. In exploring the underlying mechanism for this, we found that CXCR4, a key molecule involved in LSC/BM interactions, was specifically downregulated in CD123 overexpressing cells in a manner dependent on IL-3 signaling. CXCR4 downregulation was sufficient to alter the chemotactic response of hematopoietic cells to stromal derived factor-1 (SDF-1). Thus, we propose that the overexpression of CD123 in AML LSC dictates their location by altering CXCR4/SDF-1 interaction in the BM, raising the possibility that this mechanism underpins the egress of BM AML LSC and more mature cells into the circulation.
Collapse
|
41
|
Han L, Jorgensen JL, Brooks C, Shi C, Zhang Q, Nogueras González GM, Cavazos A, Pan R, Mu H, Wang SA, Zhou J, Ai-Atrash G, Ciurea SO, Rettig M, DiPersio JF, Cortes J, Huang X, Kantarjian HM, Andreeff M, Ravandi F, Konopleva M. Antileukemia Efficacy and Mechanisms of Action of SL-101, a Novel Anti-CD123 Antibody Conjugate, in Acute Myeloid Leukemia. Clin Cancer Res 2017; 23:3385-3395. [PMID: 28096272 DOI: 10.1158/1078-0432.ccr-16-1904] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/13/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022]
Abstract
Purpose: The persistence of leukemia stem cells (LSC)-containing cells after induction therapy may contribute to minimal residual disease (MRD) and relapse in acute myeloid leukemia (AML). We investigated the clinical relevance of CD34+CD123+ LSC-containing cells and antileukemia potency of a novel antibody conjugate SL-101 in targeting CD123+ LSCs.Experimental Methods and Results: In a retrospective study on 86 newly diagnosed AML patients, we demonstrated that a higher proportion of CD34+CD123+ LSC-containing cells in remission was associated with persistent MRD and predicted shorter relapse-free survival in patients with poor-risk cytogenetics. Using flow cytometry, we explored the potential benefit of therapeutic targeting of CD34+CD38-CD123+ cells by SL-101, a novel antibody conjugate comprising an anti-CD123 single-chain Fv fused to Pseudomonas exotoxin A The antileukemia potency of SL-101 was determined by the expression levels of CD123 antigen in a panel of AML cell lines. Colony-forming assay established that SL-101 strongly and selectively suppressed the function of leukemic progenitors while sparing normal counterparts. The internalization, protein synthesis inhibition, and flow cytometry assays revealed the mechanisms underlying the cytotoxic activities of SL-101 involved rapid and efficient internalization of antibody, sustained inhibition of protein synthesis, induction of apoptosis, and blockade of IL3-induced p-STAT5 and p-AKT signaling pathways. In a patient-derived xenograft model using NSG mice, the repopulating capacity of LSCs pretreated with SL-101 in vitro was significantly impaired.Conclusions: Our data define the mechanisms by which SL-101 targets AML and warrant further investigation of the clinical application of SL-101 and other CD123-targeting strategies in AML. Clin Cancer Res; 23(13); 3385-95. ©2017 AACR.
Collapse
Affiliation(s)
- Lina Han
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jeffrey L Jorgensen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Ce Shi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Antonio Cavazos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rongqing Pan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Mu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sa A Wang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jin Zhou
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gheath Ai-Atrash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan O Ciurea
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mike Rettig
- Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Jorge Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
42
|
Cheng G, Liu F, Asai T, Lai F, Man N, Xu H, Chen S, Greenblatt S, Hamard PJ, Ando K, Chen X, Wang L, Martinez C, Tadi M, Wang L, Xu M, Yang FC, Shiekhattar R, Nimer SD. Loss of p300 accelerates MDS-associated leukemogenesis. Leukemia 2016; 31:1382-1390. [PMID: 27881875 DOI: 10.1038/leu.2016.347] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/14/2016] [Accepted: 10/21/2016] [Indexed: 12/19/2022]
Abstract
The role that changes in DNA methylation and histone modifications have in human malignancies is poorly understood. p300 and CREB-binding protein (CBP), two distinct but highly homologous lysine acetyltransferases, are mutated in several cancers, suggesting their role as tumor suppressors. In the current study, we found that deletion of p300, but not CBP, markedly accelerated the leukemogenesis ofNup98-HoxD13 (NHD13) transgenic mice, an animal model that phenotypically copies human myelodysplastic syndrome (MDS). p300 deletion restored the ability of NHD13 expressing hematopoietic stem and progenitor cells (HSPCs) to self-renew in vitro, and to expand in vivo, with an increase in stem cell symmetric self-renewal divisions and a decrease in apoptosis. Furthermore, loss of p300, but not CBP, promoted cytokine signaling, including enhanced activation of the MAPK and JAK/STAT pathways in the HSPC compartment. Altogether, our data indicate that p300 has a pivotal role in blocking the transformation of MDS to acute myeloid leukemia, a role distinct from that of CBP.
Collapse
Affiliation(s)
- G Cheng
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - F Liu
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - T Asai
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - F Lai
- Department of Human Genetics, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - N Man
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - H Xu
- Department of Medicine, Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York,NY,USA
| | - S Chen
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - S Greenblatt
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - P-J Hamard
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - K Ando
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - X Chen
- Department of Public Health Sciences, Division of Biostatistics, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - L Wang
- Department of Public Health Sciences, Division of Biostatistics, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - C Martinez
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - M Tadi
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - L Wang
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - M Xu
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - F-C Yang
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| | - R Shiekhattar
- Department of Human Genetics, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - S D Nimer
- Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA.,Department of Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL,USA
| |
Collapse
|
43
|
|
44
|
Wei C, Wen H, Yuan L, McHale CM, Li H, Wang K, Yuan J, Yang X, Zhang L. Formaldehyde induces toxicity in mouse bone marrow and hematopoietic stem/progenitor cells and enhances benzene-induced adverse effects. Arch Toxicol 2016; 91:921-933. [PMID: 27339418 DOI: 10.1007/s00204-016-1760-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/13/2016] [Indexed: 12/13/2022]
Abstract
FA in air for 2 weeks, mimicking occupational exposure, then measured complete blood counts, nucleated BM cell count, and myeloid progenitor colony formation. We also investigated potential mechanisms of FA toxicity, including reactive oxygen species (ROS) generation, apoptosis, and hematopoietic growth factor and receptor levels. FA exposure significantly reduced nucleated BM cells and BM-derived colony-forming unit-granulocyte-macrophage (CFU-GM) and burst-forming unit-erythroid (BFU-E); down-regulated GM-CSFRα and EPOR expression; increased ROS in nucleated BM, spleen and CFU-GM cells; and increased apoptosis in nucleated spleen and CFU-GM cells. FA and BZ each similarly altered BM mature cells and stem/progenitor counts, BM and CFU-GM ROS, and apoptosis in spleen and CFU-GM but had differential effects on other end points. Co-exposure was more potent for several end points. Thus, FA is toxic to the mouse hematopoietic system, including BM stem/progenitor cells, and it enhances BZ-induced toxic effects. Our findings suggest that FA may induce BM toxicity by affecting myeloid progenitor growth and survival through oxidative damage and reduced expression levels of GM-CSFRα and EPOR.
Collapse
Affiliation(s)
- Chenxi Wei
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, 430079, Hubei, China.,Key Laboratory of Ecological Safety Monitoring and Evaluation, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Huaxiao Wen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Langyue Yuan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Cliona M McHale
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Hui Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Kun Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, 430079, Hubei, China.,Division of Biostatistics, Department of Population Health, School of Medicine, New York University, New York, NY, USA
| | - Junlin Yuan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, 430079, Hubei, China
| | - Xu Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, 430079, Hubei, China.
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
45
|
Moradi-Kalbolandi S, Davani D, Golkar M, Habibi-Anbouhi M, Abolhassani M, Shokrgozar MA. Soluble Expression and Characterization of a New scFv Directed to Human CD123. Appl Biochem Biotechnol 2016; 178:1390-406. [DOI: 10.1007/s12010-015-1954-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/07/2015] [Indexed: 12/23/2022]
|
46
|
CD123 immunostaining patterns in systemic mastocytosis: differential expression in disease subgroups and potential prognostic value. Leukemia 2015; 30:914-8. [DOI: 10.1038/leu.2015.348] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/09/2015] [Accepted: 12/07/2015] [Indexed: 12/30/2022]
|
47
|
Guo Y, Updegraff BL, Park S, Durakoglugil D, Cruz VH, Maddux S, Hwang TH, O'Donnell KA. Comprehensive Ex Vivo Transposon Mutagenesis Identifies Genes That Promote Growth Factor Independence and Leukemogenesis. Cancer Res 2015; 76:773-86. [PMID: 26676752 DOI: 10.1158/0008-5472.can-15-1697] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/18/2015] [Indexed: 11/16/2022]
Abstract
Aberrant signaling through cytokine receptors and their downstream signaling pathways is a major oncogenic mechanism underlying hematopoietic malignancies. To better understand how these pathways become pathologically activated and to potentially identify new drivers of hematopoietic cancers, we developed a high-throughput functional screening approach using ex vivo mutagenesis with the Sleeping Beauty transposon. We analyzed over 1,100 transposon-mutagenized pools of Ba/F3 cells, an IL3-dependent pro-B-cell line, which acquired cytokine independence and tumor-forming ability. Recurrent transposon insertions could be mapped to genes in the JAK/STAT and MAPK pathways, confirming the ability of this strategy to identify known oncogenic components of cytokine signaling pathways. In addition, recurrent insertions were identified in a large set of genes that have been found to be mutated in leukemia or associated with survival, but were not previously linked to the JAK/STAT or MAPK pathways nor shown to functionally contribute to leukemogenesis. Forced expression of these novel genes resulted in IL3-independent growth in vitro and tumorigenesis in vivo, validating this mutagenesis-based approach for identifying new genes that promote cytokine signaling and leukemogenesis. Therefore, our findings provide a broadly applicable approach for classifying functionally relevant genes in diverse malignancies and offer new insights into the impact of cytokine signaling on leukemia development.
Collapse
Affiliation(s)
- Yabin Guo
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Barrett L Updegraff
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sunho Park
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Deniz Durakoglugil
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Victoria H Cruz
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sarah Maddux
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tae Hyun Hwang
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kathryn A O'Donnell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas. Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
48
|
Chandran P, Pavithran K, Sidharthan N, Sasidharan A, Nair S, Koyakutty M. Protein Nanomedicine Exerts Cytotoxicity toward CD34 + CD38 - CD123 + Leukemic Stem Cells. ACS Biomater Sci Eng 2015; 1:1194-1199. [PMID: 33429669 DOI: 10.1021/acsbiomaterials.5b00361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The efficacy of protein-vorinostat nanomedicine (NV) is demonstrated in leukemic stem cells (LSC) isolated from refractory acute myeloid leukemia (AML) patient samples, where it successfully ablated both CD34+ CD38- CD123+ LSC and non-LSC "leukemic blast" compartments, without inducing myelosuppression or hemotoxicity. Besides, NV also exerted excellent synergistic lethality against leukemic bone marrow cells (BMC) at lower concentrations (0.1 μM) in combination with DNA methyltransferase (DNMT) inhibitor, decitabine. Considering the extermination of resilient LSC and synergism with decitabine, NV shows promise for clinical translation in the setting of a more tolerable and effective epigenetic targeted therapy for leukemia.
Collapse
Affiliation(s)
- Parwathy Chandran
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Keechilat Pavithran
- Department of Medical Oncology, Amrita Institute of Medical Sciences and Research Centre, Kochi, Kerala 682041, India
| | - Neeraj Sidharthan
- Department of Medical Oncology, Amrita Institute of Medical Sciences and Research Centre, Kochi, Kerala 682041, India
| | - Abhilash Sasidharan
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Shantikumar Nair
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| | - Manzoor Koyakutty
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kochi, Kerala 682041, India
| |
Collapse
|
49
|
Du W, Li J, Liu W, He Y, Yao J, Liu Y, Lin J, Zheng J. Interleukin-3 receptor α chain (CD123) is preferentially expressed in immature T-ALL and may not associate with outcomes of chemotherapy. Tumour Biol 2015; 37:3817-21. [PMID: 26474588 DOI: 10.1007/s13277-015-3272-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/17/2015] [Indexed: 12/28/2022] Open
Abstract
Interleukin-3 (IL-3) receptor α chain (CD123) plays an essential role in regulating the proliferation of hematopoietic stem cells. In the hematopoietic malignancies, CD123 expression has been found in acute myeloid leukemia (AML), B-precursor acute lymphoblastic leukemia (B-ALL), as well as dendritic cell malignancies. However, whether CD123 is also expressed in T-acute lymphoblastic leukemia (T-ALL) remains unknown. Using multi-parameter flow cytometry, we analyzed CD123 expression in 160 consecutive diagnostic T-ALL patients, including 88 pediatric T-ALL cases and 72 adult T-ALL cases. The minimal residual disease (MRD) was detected after one course of induction therapy to evaluate the treatment effects. CD123 expression was detected in 24 out of 88 (27 %) pediatric T-ALLs and 30 out of 72 (42 %) adult T-ALLs. Further analysis revealed that CD123 expression is associated with the maturation stage of T-ALLs. The frequencies of CD123-positive cases decreased from 83 to 40 % and 21 % in early T-precursor ALLs, T-precursor ALLs, and mature T-ALLs, respectively. Interestingly, we detected the CD4+CD8+ double-positive leukemic cells in 22 immature and 34 mature T-ALL patients. Of note, only 4 % of these patients expressed CD123. In addition, we found that 79 % of CD33+ and 64 % of CD117+ immature T-ALL patients also expressed CD123. However, CD123 expression did not predict the outcomes of the first course of induction therapy in T-ALL patients. In conclusion, we found that CD123 is preferentially expressed in immature T-ALL. Moreover, CD123 expression is strongly associated with cross-lineage expression of myeloid markers in early T-precursor ALL.
Collapse
Affiliation(s)
- Wen Du
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Li
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Liu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanli He
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junxia Yao
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Liu
- Wuhan Kindstar Diagnostics Co., Ltd, Wuhan, China
| | - Jun Lin
- Wuhan Kindstar Diagnostics Co., Ltd, Wuhan, China
| | - Jine Zheng
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
50
|
Pelosi E, Castelli G, Testa U. Targeting LSCs through membrane antigens selectively or preferentially expressed on these cells. Blood Cells Mol Dis 2015; 55:336-46. [PMID: 26460257 DOI: 10.1016/j.bcmd.2015.07.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 07/17/2015] [Indexed: 02/08/2023]
Abstract
Studies of xenotransplantation of bone marrow and blood cells of AML patients have supported the existence of rare leukemic stem cells, able to initiate and maintain the leukemic process and bearing the typical leukemic abnormalities. LSCs possess self-renewal capacity and are responsible for the growth of the more differentiated leukemic progeny in the bone marrow and in the blood. These cells are more resistant than bulk leukemic cells to anti-leukemic drugs, thus survive to treatment and are, at a large extent, responsible for leukemia relapse. During the last two decades, considerable progresses have been made in the understanding of the peculiar cellular and molecular properties of LSCs. In this context, particularly relevant was the discovery of several membrane markers, selectively or preferentially expressed on LSCs. These membrane markers offer now unique opportunities to identify LSCs and to distinguish them from normal HSCs, to monitor the response of the various anti-leukemic treatments at the level of the LSC compartment, to identify relevant therapeutic targets. Concerning this last point, the most promising therapeutic targets are CD33 and CD123.
Collapse
Affiliation(s)
- Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Suepriore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Suepriore di Sanità, Rome, Italy
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Suepriore di Sanità, Rome, Italy
| |
Collapse
|