1
|
Wang ZK, Zhang ZW, Lyu ZS, Xing T, Liang M, Shen MZ, Li CY, Zhang XY, Chen DD, Wang YZ, Hu LJ, Jiang H, Wang Y, Jiang Q, Zhang XH, Kong Y, Huang XJ. Inhibition of TGF-β signaling in bone marrow endothelial cells promotes hematopoietic recovery in acute myeloid leukemia patients. Cancer Lett 2024; 605:217290. [PMID: 39396705 DOI: 10.1016/j.canlet.2024.217290] [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: 09/12/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024]
Abstract
Although it is an effective treatment for acute myeloid leukemia (AML), chemotherapy leads to myelosuppression and poor hematopoietic reconstruction. Hematopoiesis is regulated by bone marrow (BM) endothelial cells (ECs), and BM ECs are dysfunctional in acute leukemia patients with poor hematopoietic reconstitution after allogenic hematopoietic stem cell transplantation. Thus, it is crucial to explore the underlying mechanism of EC impairment and establish strategies for targeted therapy. TGF-β signaling was found to be upregulated in ECs from AML patients in complete remission (CR ECs) and led to CR EC damage. Administration of a TGF-β inhibitor rescued the dysfunction of ECs caused by TGF-β1 expression in vitro, especially their hematopoiesis-supporting ability. Moreover, inhibition of TGF-β expression repaired the BM EC damage triggered by chemotherapy in both AML patients in vitro and in an AML-CR murine model, and restored normal hematopoiesis without promoting AML progression. Mechanistically, our data reveal alterations in the transcriptomic pattern of damaged BM ECs, accompanied by the overexpression of downstream molecules TGF-βR1, pSmad2/3, and functional genes related to adhesion, angiogenesis suppression and pro-apoptosis. Collectively, our findings reveal for the first time that the activation of TGF-β signaling leads to BM EC dysfunction and poor hematopoietic reconstitution. Targeting TGF-β represents a potential therapeutic strategy to promote multilineage hematopoiesis, thereby benefiting more cancer patients who suffer from myelosuppression after chemotherapy.
Collapse
Affiliation(s)
- Zhen-Kun Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Zhi-Wei Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Zhong-Shi Lyu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Tong Xing
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Mi Liang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Meng-Zhu Shen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Chen-Yuan Li
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xin-Yan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Dan-Dan Chen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Ya-Zhe Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Li-Juan Hu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Hao Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Qian Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuan Kong
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; State Key Laboratory of Natural and Biomimetic Drugs, Beijing, China.
| |
Collapse
|
2
|
Wu X, Wang F, Yang X, Gong Y, Niu T, Chu B, Qu Y, Qian Z. Advances in Drug Delivery Systems for the Treatment of Acute Myeloid Leukemia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403409. [PMID: 38934349 DOI: 10.1002/smll.202403409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Acute myeloid leukemia (AML) is a common and catastrophic hematological neoplasm with high mortality rates. Conventional therapies, including chemotherapy, hematopoietic stem cell transplantation (HSCT), immune therapy, and targeted agents, have unsatisfactory outcomes for AML patients due to drug toxicity, off-target effects, drug resistance, drug side effects, and AML relapse and refractoriness. These intrinsic limitations of current treatments have promoted the development and application of nanomedicine for more effective and safer leukemia therapy. In this review, the classification of nanoparticles applied in AML therapy, including liposomes, polymersomes, micelles, dendrimers, and inorganic nanoparticles, is reviewed. In addition, various strategies for enhancing therapeutic targetability in nanomedicine, including the use of conjugating ligands, biomimetic-nanotechnology, and bone marrow targeting, which indicates the potential to reverse drug resistance, are discussed. The application of nanomedicine for assisting immunotherapy is also involved. Finally, the advantages and possible challenges of nanomedicine for the transition from the preclinical phase to the clinical phase are discussed.
Collapse
Affiliation(s)
- Xia Wu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Fangfang Wang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Xijing Yang
- The Experimental Animal Center of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yuping Gong
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ting Niu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Bingyang Chu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ying Qu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Zhiyong Qian
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
3
|
Liu TY, Fu X, Yang Y, Gu J, Xiao M, Li DJ. Synergistic Effects of Glutamine Deprivation and Metformin in Acute Myeloid Leukemia. Curr Med Sci 2024; 44:799-808. [PMID: 39096478 DOI: 10.1007/s11596-024-2913-z] [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: 01/18/2024] [Accepted: 06/19/2024] [Indexed: 08/05/2024]
Abstract
OBJECTIVE The metabolic reprogramming of acute myeloid leukemia (AML) cells is a compensatory adaptation to meet energy requirements for rapid proliferation. This study aimed to examine the synergistic effects of glutamine deprivation and metformin exposure on AML cells. METHODS SKM-1 cells (an AML cell line) were subjected to glutamine deprivation and/or treatment with metformin or bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl) ethyl sulfide (BPTES, a glutaminase inhibitor) or cytarabine. Cell viability was detected by Cell Counting Kit-8 (CCK-8) assay, and cell apoptosis and reactive oxygen species (ROS) by flow cytometry. Western blotting was conducted to examine the levels of apoptotic proteins, including cleaved caspase-3 and poly(ADP-ribose) polymerase (PARP). Moreover, the human long noncoding RNA (lncRNA) microarray was used to analyze gene expression after glutamine deprivation, and results were confirmed with quantitative RT-PCR (qRT-PCR). The expression of metallothionein 2A (MT2A) was suppressed using siRNA. Cell growth and apoptosis were further detected by CCK-8 assay and flow cytometry, respectively, in cells with MT2A knockdown. RESULTS Glutamine deprivation or treatment with BPTES inhibited cell growth and induced apoptosis in SKM-1 cells. The lncRNA microarray result showed that the expression of MT family genes was significantly upregulated after glutamine deprivation. MT2A knockdown increased apoptosis, while proliferation was not affected in SKM-1 cells. In addition, metformin inhibited cell growth and induced apoptosis in SKM-1 cells. Both glutamine deprivation and metformin enhanced the sensitivity of SKM-1 cells to cytarabine. Furthermore, the combination of glutamine deprivation with metformin exhibited synergistic antileukemia effects on SKM-1 cells. CONCLUSION Targeting glutamine metabolism in combination with metformin is a promising new therapeutic strategy for AML.
Collapse
Affiliation(s)
- Tong-Yuan Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing Fu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia Gu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Deng-Ju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
4
|
Chang X, Qu F, Li C, Zhang J, Zhang Y, Xie Y, Fan Z, Bian J, Wang J, Li Z, Xu X. Development and therapeutic potential of GSPT1 molecular glue degraders: A medicinal chemistry perspective. Med Res Rev 2024; 44:1727-1767. [PMID: 38314926 DOI: 10.1002/med.22024] [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: 09/20/2023] [Revised: 12/18/2023] [Accepted: 01/21/2024] [Indexed: 02/07/2024]
Abstract
Unprecedented therapeutic targeting of previously undruggable proteins has now been achieved by molecular-glue-mediated proximity-induced degradation. As a small GTPase, G1 to S phase transition 1 (GSPT1) interacts with eRF1, the translation termination factor, to facilitate the process of translation termination. Studied demonstrated that GSPT1 plays a vital role in the acute myeloid leukemia (AML) and MYC-driven lung cancer. Thus, molecular glue (MG) degraders targeting GSPT1 is a novel and promising approach for treating AML and MYC-driven cancers. In this Perspective, we briefly summarize the structural and functional aspects of GSPT1, highlighting the latest advances and challenges in MG degraders, as well as some representative patents. The structure-activity relationships, mechanism of action and pharmacokinetic features of MG degraders are emphasized to provide a comprehensive compendium on the rational design of GSPT1 MG degraders. We hope to provide an updated overview, and design guide for strategies targeting GSPT1 for the treatment of cancer.
Collapse
Affiliation(s)
- Xiujin Chang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fangui Qu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chunxiao Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jingtian Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yanqing Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Xie
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhongpeng Fan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jinlei Bian
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jubo Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhiyu Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xi Xu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
5
|
Vázquez X, Lumbreras-Iglesias P, Rodicio MR, Fernández J, Bernal T, Moreno AF, de Ugarriza PL, Fernández-Verdugo A, Margolles A, Sabater C. Study of the intestinal microbiota composition and the effect of treatment with intensive chemotherapy in patients recovered from acute leukemia. Sci Rep 2024; 14:5585. [PMID: 38454103 PMCID: PMC10920697 DOI: 10.1038/s41598-024-56054-w] [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: 10/27/2023] [Accepted: 03/01/2024] [Indexed: 03/09/2024] Open
Abstract
A dataset comprising metagenomes of outpatients (n = 28) with acute leukemia (AL) and healthy controls (n = 14) was analysed to investigate the associations between gut microbiota composition and metabolic activity and AL. According to the results obtained, no significant differences in the microbial diversity between AL outpatients and healthy controls were found. However, significant differences in the abundance of specific microbial clades of healthy controls and AL outpatients were found. We found some differences at taxa level. The relative abundance of Enterobacteriaceae, Prevotellaceae and Rikenellaceae was increased in AL outpatients, while Bacteirodaceae, Bifidobacteriaceae and Lachnospiraceae was decreased. Interestingly, the abundances of several taxa including Bacteroides and Faecalibacterium species showed variations based on recovery time from the last cycle of chemotherapy. Functional annotation of metagenome-assembled genomes (MAGs) revealed the presence of functional domains corresponding to therapeutic enzymes including L-asparaginase in a wide range of genera including Prevotella, Ruminococcus, Faecalibacterium, Alistipes, Akkermansia. Metabolic network modelling revealed potential symbiotic relationships between Veillonella parvula and Levyella massiliensis and several species found in the microbiota of AL outpatients. These results may contribute to develop strategies for the recovery of microbiota composition profiles in the treatment of patients with AL.
Collapse
Grants
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- FIS PI21/01590 Fondo de Investigación Sanitaria, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Spain
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
- GRUPIN IDI/2022/000033 Regional Ministry of Science of Asturias
Collapse
Affiliation(s)
- Xenia Vázquez
- Dairy Research Institute of Asturias (IPLA), Spanish National Research Council, (CSIC), Villaviciosa, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), MicroHealth Group, Oviedo, Spain
| | - Pilar Lumbreras-Iglesias
- Traslational Microbiology Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Clinical Microbiology, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
- Department of Hematology Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), 33011, Oviedo, Spain
| | - M Rosario Rodicio
- Traslational Microbiology Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Functional Biology, Microbiology Area, University of Oviedo, Oviedo, Spain
| | - Javier Fernández
- Traslational Microbiology Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Clinical Microbiology, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
- Research & Innovation, Artificial Intelligence and Statistical Department, Pragmatech AI Solutions, Oviedo, Spain
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Madrid, Spain
| | - Teresa Bernal
- Department of Hematology Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), 33011, Oviedo, Spain
| | - Ainhoa Fernández Moreno
- Department of Hematology Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), 33011, Oviedo, Spain
| | - Paula López de Ugarriza
- Department of Hematology Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), 33011, Oviedo, Spain
| | - Ana Fernández-Verdugo
- Traslational Microbiology Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Clinical Microbiology, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Abelardo Margolles
- Dairy Research Institute of Asturias (IPLA), Spanish National Research Council, (CSIC), Villaviciosa, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), MicroHealth Group, Oviedo, Spain
| | - Carlos Sabater
- Dairy Research Institute of Asturias (IPLA), Spanish National Research Council, (CSIC), Villaviciosa, Asturias, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), MicroHealth Group, Oviedo, Spain.
| |
Collapse
|
6
|
Zhang K, Huang R, Ji M, Lin S, Lai F, Wu D, Tian H, Bi J, Peng S, Hu J, Sheng L, Li Y, Chen X, Xu H. Rational design and optimization of novel 4-methyl quinazoline derivatives as PI3K/HDAC dual inhibitors with benzamide as zinc binding moiety for the treatment of acute myeloid leukemia. Eur J Med Chem 2024; 264:116015. [PMID: 38048697 DOI: 10.1016/j.ejmech.2023.116015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/06/2023]
Abstract
Simultaneous inhibition of PI3K and HDAC has shown promise for treating various cancers, leading to discovery and development of their dual inhibitors as novel anticancer agents. Herein, we disclose a new series of PI3K/HDAC dual inhibitors bearing a benzamide moiety as the pharmacophore of HDAC inhibition. Based on systematic structure-activity relationship study, compounds 36 and 51 featuring an alkyl and benzoyl linker respectively were identified with favorable potencies against both PI3K and HDAC. In cellular assays, compounds 36 and 51 showed significantly enhanced antiproliferative activities against various cancer cell lines relative to single-target inhibitors. Furthermore, western blotting analysis shows compounds 36 and 51 suppressed AKT phosphorylation and increased H3 acetylation in MV4-11 cells, while flow cytometry analysis reveals both compounds dose-dependently induced cell cycle arrest and cell apoptosis. Supported by pharmacokinetic studies, compounds 36 and 51 were subjected to the in vivo evaluation in a MV4-11 xenograft model, demonstrating significant and dose-dependent anticancer efficacies. Overall, this work provides a promising approach for the treatment of AML by simultaneously inhibiting PI3K and HDAC with a dual inhibitor.
Collapse
Affiliation(s)
- Kehui Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Rui Huang
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, 301617, China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ming Ji
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Songwen Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Fangfang Lai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Deyu Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Hua Tian
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Jinhui Bi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Shouguo Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Jiaqi Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China
| | - Li Sheng
- Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yan Li
- Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China.
| | - Heng Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Key Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences, Beijing, 100050, China.
| |
Collapse
|
7
|
Wu B, Xu Y, Tang M, Jiang Y, Zhang T, Huang L, Wang S, Hu Y, Zhou K, Zhang X, Chen M. A Metabolome and Microbiome Analysis of Acute Myeloid Leukemia: Insights into the Carnosine-Histidine Metabolic Pathway. TOXICS 2023; 12:14. [PMID: 38250970 PMCID: PMC10821349 DOI: 10.3390/toxics12010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/17/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024]
Abstract
Metabolism underlies the pathogenesis of acute myeloid leukemia (AML) and can be influenced by gut microbiota. However, the specific metabolic changes in different tissues and the role of gut microbiota in AML remain unclear. In this study, we analyzed the metabolome differences in blood samples from patients with AML and healthy controls using UPLC-Q-Exactive. Additionally, we examined the serum, liver, and fecal metabolome of AML model mice and control mice using UPLC-Q-Exactive. The gut microbiota of the mice were analyzed using 16S rRNA sequencing. Our UPLC-MS analysis revealed significant differences in metabolites between the AML and control groups in multiple tissue samples. Through cross-species validation in humans and animals, as well as reverse validation of Celastrol, we discovered that the Carnosine-Histidine metabolic pathway may play a potential role in the occurrence and progression of AML. Furthermore, our analysis of gut microbiota showed no significant diversity changes, but we observed a significant negative correlation between the key metabolite Carnosine and Peptococcaceae and Campylobacteraceae. In conclusion, the Carnosine-Histidine metabolic pathway influences the occurrence and progression of AML, while the gut microbiota might play a role in this process.
Collapse
Affiliation(s)
- Binxiong Wu
- Department of Hygienic Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing 211166, China;
| | - Yuntian Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (Y.X.); (M.T.); (Y.J.); (L.H.); (S.W.); (K.Z.)
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Miaomiao Tang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (Y.X.); (M.T.); (Y.J.); (L.H.); (S.W.); (K.Z.)
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yingtong Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (Y.X.); (M.T.); (Y.J.); (L.H.); (S.W.); (K.Z.)
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ting Zhang
- Women’s Hospital of Jiangnan University, Wuxi 214002, China;
| | - Lei Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (Y.X.); (M.T.); (Y.J.); (L.H.); (S.W.); (K.Z.)
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Shuyang Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (Y.X.); (M.T.); (Y.J.); (L.H.); (S.W.); (K.Z.)
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yanhui Hu
- Sir Run Run Hospital of Nanjing Medical University, Nanjing 211166, China;
| | - Kun Zhou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (Y.X.); (M.T.); (Y.J.); (L.H.); (S.W.); (K.Z.)
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoling Zhang
- Department of Hygienic Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing 211166, China;
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (Y.X.); (M.T.); (Y.J.); (L.H.); (S.W.); (K.Z.)
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
8
|
Chen PY, Lin CY, Wu CL, Keak PY, Liou JW, Gao WY, Lin LI, Yen JH. Pinostrobin modulates FOXO3 expression, nuclear localization, and exerts antileukemic effects in AML cells and zebrafish xenografts. Chem Biol Interact 2023; 385:110729. [PMID: 37777166 DOI: 10.1016/j.cbi.2023.110729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/30/2023] [Accepted: 09/21/2023] [Indexed: 10/02/2023]
Abstract
Acute myeloid leukemia (AML) is a disease characterized by abnormal cell proliferation in the bone marrow and is the most common quickly progressive leukemia in adults. Pinostrobin, a flavonoid phytochemical, has been reported to exhibit antioxidant, anti-inflammatory, and anticancer properties. In this study, we aimed to investigate the antileukemic effects of pinostrobin and its molecular mechanisms in human AML cells. Our study found that pinostrobin (0-80 μM) significantly reduced the viability of human AML cells, with the pronounced cytotoxic effects observed in MV4-11 > MOLM-13 > HL-60 > U-937 > THP-1 cells. Pinostrobin was found to suppress leukemia cell proliferation, modulate cell cycle progression, promote cell apoptosis, and induce monocytic differentiation in MV4-11 cells. In animal studies, pinostrobin significantly suppressed the growth of leukemia cells in a zebrafish xenograft model. Microarray-based transcriptome analysis showed that the differentially expressed genes (DEGs) in pinostrobin-treated cells were strongly associated with enriched Gene Ontology (GO) terms related to apoptotic process, cell death, cell differentiation, cell cycle progression, and cell division. Combining DisGeNET and STRING database analysis revealed that pinostrobin upregulates forkhead box 3 (FOXO3), a tumor suppressor in cancer development, and plays an essential role in controlling AML cell viability. Our study demonstrated that pinostrobin increases FOXO3 gene expression and promotes its nuclear translocation, leading to the inhibition of cell growth. Finally, the study found that pinostrobin, when combined with cytarabine, synergistically reduces the viability of AML cells. Our current findings shed light on pinostrobin's mechanisms in inhibiting leukemia cell growth, highlighting its potential as a chemotherapeutic agent or nutraceutical supplement for AML prevention or treatment.
Collapse
Affiliation(s)
- Pei-Yi Chen
- Laboratory of Medical Genetics, Genetic Counseling Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970374, Taiwan; Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, 970374, Taiwan
| | - Ching-Yen Lin
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, 970374, Taiwan
| | - Chia-Ling Wu
- Laboratory of Medical Genetics, Genetic Counseling Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970374, Taiwan
| | - Pei Ying Keak
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, 970374, Taiwan
| | - Je-Wen Liou
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 970374, Taiwan
| | - Wan-Yun Gao
- Institute of Medical Sciences, Tzu Chi University, Hualien, 970374, Taiwan
| | - Liang-In Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 10048, Taiwan
| | - Jui-Hung Yen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, 970374, Taiwan; Institute of Medical Sciences, Tzu Chi University, Hualien, 970374, Taiwan.
| |
Collapse
|
9
|
Yuan H, Lu Y, Feng Y, Wang N. Epigenetic inhibitors for cancer treatment. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 383:89-144. [PMID: 38359972 DOI: 10.1016/bs.ircmb.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Epigenetics is a heritable and reversible modification that occurs independent of the alteration of primary DNA sequence but remarkably affects genetic expression. Aberrant epigenetic regulators are frequently observed in cancer progression not only influencing the behavior of tumor cells but also the tumor-associated microenvironment (TME). Increasing evidence has shown their great potential as biomarkers to predict clinical outcomes and chemoresistance. Hence, targeting the deregulated epigenetic regulators would be a compelling strategy for cancer treatment. So far, current epigenetic drugs have shown promising efficacy in both preclinical trials and clinical treatment of cancer, which encourages research discoveries on the development of novel epigenetic inhibitors either from natural compounds or artificial synthesis. However, only a few have been approved by the FDA, and more effort needs to be put into the related research. This chapter will update the applications and latest progress of epigenetic inhibitors in cancer treatment and provide prospects for the future development of epigenetic drugs.
Collapse
Affiliation(s)
- Hongchao Yuan
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yuanjun Lu
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
10
|
Xie J, Zhao X, Zhang P, Zhang Y, Cheng R, Zhong Z, Deng C. Codelivery of BCL2 and MCL1 Inhibitors Enabled by Phenylboronic Acid-Functionalized Polypeptide Nanovehicles for Synergetic and Potent Therapy of Acute Myeloid Leukemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204866. [PMID: 36683178 PMCID: PMC10015845 DOI: 10.1002/advs.202204866] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Acute myeloid leukemia (AML) is the most refractory hematologic malignancy characterized by acute onset, rapid progression, and high recurrence rate. Here, codelivery of BCL2 (ABT199) and MCL1 (TW37) inhibitors using phenylboronic acid-functionalized polypeptide nanovehicles to achieve synergetic and potent treatment of AML is adopted. Leveraging the dynamic boronic ester bonds, BN coordination, and π-π stacking, the nanovehicles reveal remarkably efficient and robust drug coencapsulation. ABT199 can induce a series of pro-apoptotic reactions by promoting the dissociation of the pro-apoptotic protein Bim from BCL2, while the released Bim is often captured by MCL1 protein overexpressed in AML. TW37 has a strong inhibitory ability to MCL1, thereby can restrain the depletion of Bim protein. Dual inhibitor-loaded nanoparticles (NPAT) reveal excellent stability, acid/enzyme/H2 O2 -triggered drug release, and significant cytotoxicity toward MOLM-13-Luc and MV-411 AML cells with low half maximal inhibitory concentrations of 1.15 and 7.45 ng mL-1 , respectively. In mice bearing MOLM-13-Luc or MV-411 AML cancer, NPAT reveal significant inhibition of tumor cell infiltration in bone marrow and main organs, potent suppression of tumor growth, and remarkably elevated mouse survival. With facile construction, varying drug combination, superior safety, synergetic efficacy, the phenylboronic acid-functionalized smart nanodrugs hold remarkable potential for AML treatment.
Collapse
Affiliation(s)
- Jiguo Xie
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Xiaofei Zhao
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Peng Zhang
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Yueyue Zhang
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Ru Cheng
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| | - Chao Deng
- Biomedical Polymers Laboratoryand Jiangsu Key Laboratory of Advanced Functional Polymer Design and ApplicationCollege of ChemistryChemical Engineering and Materials Scienceand State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhou215123P. R. China
| |
Collapse
|
11
|
Ran F, Liu Y, Zhu J, Wu H, Tao W, Xie X, Hu Y, Zhang Y, Ling Y. Design, synthesis and pharmacological characterization of aminopyrimidine derivatives as BTK/FLT3 dual-target inhibitors against acute myeloid leukemia. Bioorg Chem 2023; 134:106479. [PMID: 36989958 DOI: 10.1016/j.bioorg.2023.106479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/17/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023]
Abstract
A novel class of aminopyrimidine-based Bruton's tyrosine kinase (BTK) and FMS-like tyrosine kinase 3 (FLT3) dual-target inhibitors based on the BTK inhibitor spebrutinib was designed for the treatment of acute myeloid leukemia. Representative compounds 14d, 14g, 14j and 14m effectively inhibited BTK, FLT3, and FLT3(D835Y) mutant activities with low nanomolar IC50's. These compounds displayed potent antiproliferative activities against leukemia cells with IC50's of 0.29-950 nM. In particular, 14m had IC50 values 101-1045 times lower than those of spebrutinib against all cancer cell lines tested. Compound 14m effectively induced autophagy and apoptosis in MV-4-11 cells through regulating related proteins in a dose-dependent manner. Finally, intraperitoneal administration of 14m at 20 mg/kg significantly repressed the growth of MV-4-11 cells with a TGI value of 95.68% with no apparent toxicity. These BTK/FLT3 dual-target inhibitors represent promising leads for further structural optimization and antitumor mechanism studies.
Collapse
|
12
|
Tsuzuki H, Kawase T, Nakazawa T, Mori M, Yoshida T. Anti-tumor effect of antibody drug conjugate ASP1235 targeting Fms-like tyrosine kinase 3 with venetoclax plus azacitidine in an acute myeloid leukemia xenograft mouse model. Oncotarget 2022; 13:1359-1368. [PMID: 36537913 PMCID: PMC9765856 DOI: 10.18632/oncotarget.28331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Antibody drug conjugates (ADC) are one of the attractive modalities for the treatment of acute myeloid leukemia (AML). Previously, we have developed ASP1235, a novel ADC targeting Fms-like tyrosine kinase 3 (FLT3) which is widely expressed on the leukemic blasts of AML patients. In this study, we sought to evaluate the therapeutic effect of ASP1235 in combination with venetoclax plus azacitidine, a novel standard-of-care treatment for elderly AML patients, in ASP1235 poor sensitive AML cells. To identify the suitable preclinical model, we first evaluated the growth inhibitory effect of ASP1235 on several leukemia cell lines expressing FLT3 and found that THP-1 cells were partially sensitive to ASP1235 in vitro. Furthermore, ASP1235 showed marginal anti-tumor activity in a THP-1 xenograft model. Compared to the leukemic blasts in most of the relapsed or refractory (R/R) AML patients tested, THP-1 cells expressed equivalent protein levels of Bcl-2, suggesting that ASP1235 in combination with venetoclax plus azacitidine is a rational treatment in the THP-1 model. In vitro, ASP1235 showed a cytotoxic effect on THP-1 cells in combination with venetoclax, and the combination effect was greater than the additive effect. Furthermore, ASP1235 also showed a combination effect with venetoclax plus azacitidine treatment. Similarly, the combination of ASP1235, venetoclax and azacitidine showed a superior anti-tumor effect in a THP-1 xenograft model without obvious body weight loss. These findings provide supportive evidence that the triple combination of ASP1235, venetoclax and azacitidine would improve the clinical outcome of ASP1235 monotherapy and venetoclax plus azacitidine regimen in AML patients.
Collapse
Affiliation(s)
- Hirofumi Tsuzuki
- 1Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan,Correspondence to:Hirofumi Tsuzuki, email:
| | - Tatsuya Kawase
- 1Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Taisuke Nakazawa
- 1Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Masamichi Mori
- 2Applied Research and Operations, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Taku Yoshida
- 1Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| |
Collapse
|
13
|
Zhang M, Ge Y, Xu S, Fang X, Meng J, Yu L, Wang C, Liu J, Wen T, Yang Y, Wang C, Xu H. Nanomicelles co-loading CXCR4 antagonist and doxorubicin combat the refractory acute myeloid leukemia. Pharmacol Res 2022; 185:106503. [DOI: 10.1016/j.phrs.2022.106503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/27/2022] [Accepted: 10/09/2022] [Indexed: 10/31/2022]
|
14
|
Dong Q, Xiu Y, Wang Y, Hodgson C, Borcherding N, Jordan C, Buchanan J, Taylor E, Wagner B, Leidinger M, Holman C, Thiele DJ, O’Brien S, Xue HH, Zhao J, Li Q, Meyerson H, Boyce BF, Zhao C. HSF1 is a driver of leukemia stem cell self-renewal in acute myeloid leukemia. Nat Commun 2022; 13:6107. [PMID: 36245043 PMCID: PMC9573868 DOI: 10.1038/s41467-022-33861-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 10/05/2022] [Indexed: 01/25/2023] Open
Abstract
Acute myeloid leukemia (AML) is maintained by self-renewing leukemic stem cells (LSCs). A fundamental problem in treating AML is that conventional therapy fails to eliminate LSCs, which can reinitiate leukemia. Heat shock transcription factor 1 (HSF1), a central regulator of the stress response, has emerged as an important target in cancer therapy. Using genetic Hsf1 deletion and a direct HSF1 small molecule inhibitor, we show that HSF1 is specifically required for the maintenance of AML, while sparing steady-state and stressed hematopoiesis. Mechanistically, deletion of Hsf1 dysregulates multifaceted genes involved in LSC stemness and suppresses mitochondrial oxidative phosphorylation through downregulation of succinate dehydrogenase C (SDHC), a direct HSF1 target. Forced expression of SDHC largely restores the Hsf1 ablation-induced AML developmental defect. Importantly, the growth and engraftment of human AML cells are suppressed by HSF1 inhibition. Our data provide a rationale for developing efficacious small molecules to specifically target HSF1 in AML.
Collapse
Affiliation(s)
- Qianze Dong
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA
| | - Yan Xiu
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA ,grid.410349.b0000 0004 5912 6484Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106 USA
| | - Yang Wang
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA
| | | | - Nick Borcherding
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Craig Jordan
- grid.241116.10000000107903411Division of Hematology, University of Colorado Anschutz Campus, Denver, CO 80045 USA
| | - Jane Buchanan
- grid.214572.70000 0004 1936 8294Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52240 USA
| | - Eric Taylor
- grid.214572.70000 0004 1936 8294Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52240 USA
| | - Brett Wagner
- grid.214572.70000 0004 1936 8294Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242 USA
| | - Mariah Leidinger
- grid.214572.70000 0004 1936 8294Department of Pathology, University of Iowa, Iowa City, IA 52242 USA
| | - Carol Holman
- grid.214572.70000 0004 1936 8294Department of Pathology, University of Iowa, Iowa City, IA 52242 USA
| | | | | | - Hai-hui Xue
- grid.239835.60000 0004 0407 6328Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110 USA
| | - Jinming Zhao
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA ,grid.412449.e0000 0000 9678 1884Department of Pathology, China Medical University, 77 Puhe Rd, Shenbei Xinqu, Shenyang Shi, 110122 Liaoning Sheng China
| | - Qingchang Li
- grid.412449.e0000 0000 9678 1884Department of Pathology, China Medical University, 77 Puhe Rd, Shenbei Xinqu, Shenyang Shi, 110122 Liaoning Sheng China
| | - Howard Meyerson
- grid.443867.a0000 0000 9149 4843Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 USA
| | - Brendan F. Boyce
- grid.412750.50000 0004 1936 9166Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Chen Zhao
- grid.67105.350000 0001 2164 3847Department of Pathology, Case Western Reserve University, Cleveland, OH 44106 USA ,grid.410349.b0000 0004 5912 6484Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106 USA ,grid.443867.a0000 0000 9149 4843Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 USA
| |
Collapse
|
15
|
Huang W, Sun G, Wang Q, Long Z. The research progress of targeted therapy in acute myeloid leukemia based on bibliometric analysis. Front Oncol 2022; 12:957370. [PMID: 36119476 PMCID: PMC9481238 DOI: 10.3389/fonc.2022.957370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Affiliation(s)
- Wanxue Huang
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China
| | - Gongrui Sun
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qi Wang
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China
| | - Zhiguo Long
- Department of Hematology, Fudan University Affiliated Pudong Medical Center, Shanghai, China
- *Correspondence: Zhiguo Long,
| |
Collapse
|
16
|
Zhang S, Liu J, Lu ZY, Xue YT, Mu XR, Liu Y, Cao J, Li ZY, Li F, Xu KL, Wu QY. Combination of RSK inhibitor LJH-685 and FLT3 inhibitor FF-10101 promoted apoptosis and proliferation inhibition of AML cell lines. Cell Oncol (Dordr) 2022; 45:1005-1018. [PMID: 36036884 DOI: 10.1007/s13402-022-00703-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2022] [Indexed: 11/03/2022] Open
Abstract
PURPOSE FLT3 mutations occurred in approximately one third of patients with acute myeloid leukemia (AML). FLT3-ITD mutations caused the constitutive activation of the RAS/MAPK signaling pathway. Ribosomal S6 Kinases (RSKs) were serine/threonine kinases that function downstream of the Ras/Raf/MEK/ERK signaling pathway. However, roles and mechanisms of RSKs inhibitor LJH-685, and combinational effects of LJH-685 and FLT3 inhibitor FF-10101 on AML cells were till unclear. METHODS Cell viability assay, CFSE assay, RT-qPCR, Colony formation assay, PI stain, Annexin-V/7-AAD double stain, Western blot, and Xenogeneic transplantation methods were used to used to investigate roles and mechanisms of LJH-685 in the leukemogenesis of AML. RESULTS LJH-685 inhibited the proliferation and clone formation of AML cells, caused cell cycle arrest and induced the apoptosis of AML cells via inhibiting the RSK-YB-1 signaling pathway. MV4-11 and MOLM-13 cells carrying FLT3-ITD mutations were more sensitive to LJH-685 than that of other AML cell lines. Further studies suggested that LJH-685 combined with Daunorubicin or FF- 10101 synergistically inhibited the cell viability, promoted the apoptosis and caused cycle arrest of AML cells carrying FLT3-ITD mutations. Moreover, in vivo experiments also indicated that LJH-685 combined with FF-10101 or Daunorubicin prolonged the survival time of NSG mice and reduced the leukemogenesis of AML. CONCLUSION Thus, these observations demonstrated combination of RSK inhibitor LJH-685 and FLT3 inhibitor FF-10101 showed synergism anti-leukemia effects in AML cell lines with FLT3-ITD mutations via inhibiting MAPK-RSKs-YB-1 pathway and provided new targets for therapeutic intervention especially for AML with FLT3-ITD mutations and Daunorubicin-resistant AML.
Collapse
Affiliation(s)
- Sen Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jun Liu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zi-Yi Lu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu-Tong Xue
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xing-Ru Mu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Liu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiang Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhen-Yu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Feng Li
- Department of Cell Biology and Neurobiology, Xuzhou Medical University, Xuzhou, 221002, People's Republic of China.
| | - Kai-Lin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Qing-Yun Wu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
17
|
Chen PY, Wang CY, Tsao EC, Chen YT, Wu MJ, Ho CT, Yen JH. 5-Demethylnobiletin Inhibits Cell Proliferation, Downregulates ID1 Expression, Modulates the NF-κB/TNF-α Pathway and Exerts Antileukemic Effects in AML Cells. Int J Mol Sci 2022; 23:ijms23137392. [PMID: 35806401 PMCID: PMC9266321 DOI: 10.3390/ijms23137392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is characterized by the dysregulation of hematopoietic cell proliferation, resulting in the accumulation of immature myeloid cells in bone marrow. 5-Demethylnobiletin (5-demethyl NOB), a citrus 5-hydroxylated polymethoxyflavone, has been reported to exhibit various bioactivities, such as antioxidant, anti-inflammatory and anticancer properties. In this study, we investigated the antileukemic effects of 5-demethyl NOB and its underlying molecular mechanisms in human AML cells. We found that 5-demethyl NOB (20−80 μM) significantly reduced human leukemia cell viability, and the following trend of effectiveness was observed: THP-1 ≈ U-937 > HEL > HL-60 > K562 cells. 5-Demethyl NOB (20 and 40 μM) modulated the cell cycle through the regulation of p21, cyclin E1 and cyclin A1 expression and induced S phase arrest. 5-Demethyl NOB also promoted leukemia cell apoptosis and differentiation. Microarray-based transcriptome, Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) of differentially expressed genes (DEGs) analysis showed that the expression of inhibitor of differentiation/DNA binding 1 (ID1), a gene associated with the GO biological process (BP) cell population proliferation (GO: 0008283), was most strongly suppressed by 5-demethyl NOB (40 μM) in THP-1 cells. We further demonstrated that 5-demethyl NOB-induced ID1 reduction was associated with the inhibition of leukemia cell growth. Moreover, DEGs involved in the hallmark gene set NF-κB/TNF-α signaling pathway were markedly enriched and downregulated by 5-demethyl NOB. Finally, we demonstrated that 5-demethyl NOB (20 and 40 μM), combined with cytarabine, synergistically reduced THP-1 and U-937 cell viability. Our current findings support that 5-demethyl NOB dramatically suppresses leukemia cell proliferation and may serve as a potential phytochemical for human AML chemotherapy.
Collapse
Affiliation(s)
- Pei-Yi Chen
- Center of Medical Genetics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan;
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (E.-C.T.); (Y.-T.C.)
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 11031, Taiwan
| | - En-Ci Tsao
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (E.-C.T.); (Y.-T.C.)
| | - Yu-Ting Chen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (E.-C.T.); (Y.-T.C.)
| | - Ming-Jiuan Wu
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan;
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA;
| | - Jui-Hung Yen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan; (E.-C.T.); (Y.-T.C.)
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
- Correspondence: ; Tel.: +886-3-856-5301 (ext. 2683)
| |
Collapse
|
18
|
Dunsmore L, Navo CD, Becher J, de Montes EG, Guerreiro A, Hoyt E, Brown L, Zelenay V, Mikutis S, Cooper J, Barbieri I, Lawrinowitz S, Siouve E, Martin E, Ruivo PR, Rodrigues T, da Cruz FP, Werz O, Vassiliou G, Ravn P, Jiménez-Osés G, Bernardes GJL. Controlled masking and targeted release of redox-cycling ortho-quinones via a C-C bond-cleaving 1,6-elimination. Nat Chem 2022; 14:754-765. [PMID: 35764792 PMCID: PMC9252919 DOI: 10.1038/s41557-022-00964-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/03/2022] [Indexed: 12/15/2022]
Abstract
Natural products that contain ortho-quinones show great potential as anticancer agents but have been largely discarded from clinical development because their redox-cycling behaviour results in general systemic toxicity. Here we report conjugation of ortho-quinones to a carrier, which simultaneously masks their underlying redox activity. C-benzylation at a quinone carbonyl forms a redox-inactive benzyl ketol. Upon a specific enzymatic trigger, an acid-promoted, self-immolative C-C bond-cleaving 1,6-elimination mechanism releases the redox-active hydroquinone inside cells. By using a 5-lipoxygenase modulator, β-lapachone, we created cathepsin-B-cleavable quinone prodrugs. We applied the strategy for intracellular release of β-lapachone upon antibody-mediated delivery. Conjugation of protected β-lapachone to Gem-IgG1 antibodies, which contain the variable region of gemtuzumab, results in homogeneous, systemically non-toxic and conditionally stable CD33+-specific antibody-drug conjugates with in vivo efficacy against a xenograft murine model of acute myeloid leukaemia. This protection strategy could allow the use of previously overlooked natural products as anticancer agents, thus extending the range of drugs available for next-generation targeted therapeutics.
Collapse
Affiliation(s)
- Lavinia Dunsmore
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Claudio D Navo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio-Bizkaia, Spain
| | - Julie Becher
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | | | - Ana Guerreiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Emily Hoyt
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Libby Brown
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
| | | | - Sigitas Mikutis
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Jonathan Cooper
- Wellcome-MRC Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, UK
| | - Isaia Barbieri
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Stefanie Lawrinowitz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Elise Siouve
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Esther Martin
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Pedro R Ruivo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Tiago Rodrigues
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Filipa P da Cruz
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - George Vassiliou
- Wellcome-MRC Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, UK
| | - Peter Ravn
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
- Department of Biotherapeutic Discovery, H. Lundbeck A/S, Valby, Denmark
| | - Gonzalo Jiménez-Osés
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio-Bizkaia, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| | - Gonçalo J L Bernardes
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
19
|
Gatua M, Navari M, Ong’ondi M, Onyango N, Kaggia S, Rogena E, Visani G, Abinya NA, Piccaluga PP. Molecular Profiling of Kenyan Acute Myeloid Leukemia Patients. Front Genet 2022; 13:843705. [PMID: 35836575 PMCID: PMC9274457 DOI: 10.3389/fgene.2022.843705] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 05/13/2022] [Indexed: 11/28/2022] Open
Abstract
Acute myeloid leukemia (AML) is an infrequent disease, and it is associated with high morbidity and mortality. It harbors a unique configuration of cytogenetic abnormalities and molecular mutations that can be detected using microscopic and molecular methods respectively. These genetic tests are core elements of diagnosis and prognostication in high-income countries. They are routinely incorporated in clinical decision making, allowing for the individualization of therapy. However, these tests are largely inaccessible to most patients in Kenya and therefore no data has been reported on this group of patients. The main purpose of this study is to describe the cytogenetic and molecular abnormalities of acute myeloid leukemia patients seen at the hemato-oncology unit of Kenyatta National Hospital. A cross-sectional descriptive study was carried out over a 3-month period on ten patients with a diagnosis of AML. Social demographics and clinical data were collected through a study proforma. A peripheral blood sample was collected for conventional metaphase G-banding technique and next generation sequencing. Particularly, targeted DNA sequencing (Illumina myeloid panel) and whole exome sequencing (WES) were performed. Cytogenetic analysis failed in 10/10 cases. Targeted sequencing was successfully obtained in 8 cases, whereas WES in 7. Cytogenetic studies yielded no results. There were 20 mutations detected across 10 commonly mutated genes. All patients had at least one clinically relevant mutation. Based on ELN criteria, NGS identified three patients with high-risk mutations, affecting TP53 (n = 2) and RUNX1 (n = 1). One patient was classified as favorable (PML-RARA) while 4 were standard risk. However, WT1 mutations associated with unfavorable prognosis were recorded in additional 2 cases. WES showed concordant results with targeted sequencing while unveiling more mutations that warrant further attention. In conclusion, we provide the first molecular profiling study of AML patients in Kenya including application of advanced next generation sequencing technologies, highlighting current limitations of AML diagnostics and treatment while confirming the relevance of NGS in AML characterization.
Collapse
Affiliation(s)
- Mercy Gatua
- Biobank of Research, IRCCS S. Orsola-Malpighi Academic Hospital, Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology and Medical Oncology “L. and A. Seràgnoli”, University of Bologna School of Medicine, Bologna, Italy
- Kenyatta National Hospital, Nairobi, Kenya
| | - Mohsen Navari
- Department of Medical Biotechnology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | | | - Noel Onyango
- Nairobi Hospital, University of Nairobi, Nairobi, Kenya
| | - Serah Kaggia
- Department of Pathology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Juja, Kenya
| | - Emily Rogena
- Department of Pathology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Juja, Kenya
| | - Giuseppe Visani
- Hematology and Stem Cell Transplantation, AORMN, Pesaro, Italy
| | | | - Pier Paolo Piccaluga
- Biobank of Research, IRCCS S. Orsola-Malpighi Academic Hospital, Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology and Medical Oncology “L. and A. Seràgnoli”, University of Bologna School of Medicine, Bologna, Italy
- Department of Pathology, School of Medicine, Jomo Kenyatta University of Agriculture and Technology, Juja, Kenya
- Istituto Euro-Mediterraneo di Scienza e Tecnologia (IEMEST), Palermo, Italy
- Nanchang University, Nanchang, China
- *Correspondence: Pier Paolo Piccaluga,
| |
Collapse
|
20
|
Ci T, Zhang W, Qiao Y, Li H, Zang J, Li H, Feng N, Gu Z. Delivery strategies in treatments of leukemia. Chem Soc Rev 2022; 51:2121-2144. [PMID: 35188506 DOI: 10.1039/d1cs00755f] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Leukemia is a hematological malignancy associated with the uncontrolled proliferation of mutant progenitors, suppressing the production of normal blood cells. Current treatments, including chemotherapy, radiotherapy, and immunotherapy, still lead to unsatisfactory results with a 5 year survival rate of only 30-50%. The poor prognosis is related to both disease relapse and treatment-associated toxicity. Delivery strategies can improve the in vivo pharmacokinetics of drugs, navigating the therapeutics to target cells or the tumor microenvironment and reversing drug resistance, which maximizes tumor elimination and alleviates systematic adverse effects. This review discusses available FDA-approved anti-leukemia drugs and therapies with a focus on the advances in the development of anti-leukemia drug delivery systems. Additionally, challenges in clinical translation of the delivery strategies and future research opportunities in leukemia treatment are also included.
Collapse
Affiliation(s)
- Tianyuan Ci
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Wentao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yingyu Qiao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Huangjuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province, 210009, China
| | - Jing Zang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hongjun Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Zhen Gu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China. .,Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
21
|
Sun X, Wang G, Zuo S, Niu Q, Chen X, Feng X. Preclinical Evaluation of CD64 As a Potential Target For CAR-T-cell Therapy For Acute Myeloid Leukemia. J Immunother 2022; 45:67-77. [PMID: 34864808 DOI: 10.1097/cji.0000000000000406] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/28/2021] [Indexed: 11/25/2022]
Abstract
The relapsed and refractory acute myeloid leukemia (AML) patients receiving traditional chemotherapies have poor survival rate. Chimeric antigen receptor (CAR)-modified T cells have demonstrated remarkable effectiveness against some malignancies. However, most of CAR-Ts targeting the candidate proteins on AML cells induce hematopoietic cell suppression. Because of extensive heterogeneity among different types of AML, it is essential to expand the choice of target antigen for the CAR-T treatment of AML. CD64 (FcγRI) is a transmembrane protein with broad expression on various types of AML cells, especially monocytic AML cells, but it is absent on hematopoietic stem cells (HSCs) and most of nonmonocytes. Here, we found that some types of AML patients showed the homogeneous high-level expression of CD64. So, we created a CAR-T targeting CD64 (64bbz) and further verified its high efficiency for eradicating CD64+AML cells. In addition, 64bbz showed no cytotoxicity to HSCs. Overall, we developed a new treatment option for AML by using CD64 CAR-T cells while avoiding ablation of HSCs.
Collapse
Affiliation(s)
- Xiaolei Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
| | - Guoling Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
| | - Shiyu Zuo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
| | - Qing Niu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
| | - Xiaoli Chen
- Central Laboratory, Ganzhou Key Laboratory of Molecular Medicine, the Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou
| |
Collapse
|
22
|
Innate Immune Mechanisms and Immunotherapy of Myeloid Malignancies. Biomedicines 2021; 9:biomedicines9111631. [PMID: 34829860 PMCID: PMC8615731 DOI: 10.3390/biomedicines9111631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Similar to other cancers, myeloid malignancies are thought to subvert the immune system during their development. This subversion occurs via both malignant cell-autonomous and non-autonomous mechanisms and involves manipulation of the innate and adaptive immune systems. Multiple strategies are being studied to rejuvenate, redirect, or re-enforce the immune system in order to fight off myeloid malignancies. So far, the most successful strategies include interferon treatment and antibody-based therapies, though chimeric antigen receptor (CAR) cells and immune checkpoint inhibitors are also promising therapies. In this review, we discuss the inherent immune mechanisms of defense against myeloid malignancies, currently-approved agents, and agents under investigation. Overall, we evaluate the efficacy and potential of immuno-oncology in the treatment of myeloid malignancies.
Collapse
|
23
|
Sidhu I, Barwe SP, Pillai RK, Gopalakrishnapillai A. Harnessing the Power of Induced Pluripotent Stem Cells and Gene Editing Technology: Therapeutic Implications in Hematological Malignancies. Cells 2021; 10:2698. [PMID: 34685678 PMCID: PMC8534597 DOI: 10.3390/cells10102698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/16/2022] Open
Abstract
In vitro modeling of hematological malignancies not only provides insights into the influence of genetic aberrations on cellular and molecular mechanisms involved in disease progression but also aids development and evaluation of therapeutic agents. Owing to their self-renewal and differentiation capacity, induced pluripotent stem cells (iPSCs) have emerged as a potential source of short in supply disease-specific human cells of the hematopoietic lineage. Patient-derived iPSCs can recapitulate the disease severity and spectrum of prognosis dictated by the genetic variation among patients and can be used for drug screening and studying clonal evolution. However, this approach lacks the ability to model the early phases of the disease leading to cancer. The advent of genetic editing technology has promoted the generation of precise isogenic iPSC disease models to address questions regarding the underlying genetic mechanism of disease initiation and progression. In this review, we discuss the use of iPSC disease modeling in hematological diseases, where there is lack of patient sample availability and/or difficulty of engraftment to generate animal models. Furthermore, we describe the power of combining iPSC and precise gene editing to elucidate the underlying mechanism of initiation and progression of various hematological malignancies. Finally, we discuss the power of iPSC disease modeling in developing and testing novel therapies in a high throughput setting.
Collapse
Affiliation(s)
- Ishnoor Sidhu
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| | - Sonali P. Barwe
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| | - Raju K. Pillai
- National Medical Center, Department of Pathology, City of Hope, Duarte, CA 91105, USA;
| | - Anilkumar Gopalakrishnapillai
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, Nemours Children’s Health, Wilmington, DE 19803, USA; (I.S.); (S.P.B.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
24
|
Genomic Abnormalities as Biomarkers and Therapeutic Targets in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13205055. [PMID: 34680203 PMCID: PMC8533805 DOI: 10.3390/cancers13205055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary AML is a heterogenous malignancy with a variety of underlying genomic abnormalities. Some of the genetic aberrations in AML have led to the development of specific inhibitors which were approved by the Food and Drug Administration (FDA) and are currently used to treat eligible patients. In this review, we describe five gene mutations for which approved inhibitors have been developed, the response of AML patients to these inhibitors, and the known mechanism(s) of resistance. This review also highlights the significance of developing function-based screens for target discovery in the era of personalized medicine. Abstract Acute myeloid leukemia (AML) is a highly heterogeneous malignancy characterized by the clonal expansion of myeloid stem and progenitor cells in the bone marrow, peripheral blood, and other tissues. AML results from the acquisition of gene mutations or chromosomal abnormalities that induce proliferation or block differentiation of hematopoietic progenitors. A combination of cytogenetic profiling and gene mutation analyses are essential for the proper diagnosis, classification, prognosis, and treatment of AML. In the present review, we provide a summary of genomic abnormalities in AML that have emerged as both markers of disease and therapeutic targets. We discuss the abnormalities of RARA, FLT3, BCL2, IDH1, and IDH2, their significance as therapeutic targets in AML, and how various mechanisms cause resistance to the currently FDA-approved inhibitors. We also discuss the limitations of current genomic approaches for producing a comprehensive picture of the activated signaling pathways at diagnosis or at relapse in AML patients, and how innovative technologies combining genomic and functional methods will improve the discovery of novel therapeutic targets in AML. The ultimate goal is to optimize a personalized medicine approach for AML patients and possibly those with other types of cancers.
Collapse
|
25
|
Liang C, Zhao Y, Chen C, Huang S, Deng T, Zeng X, Tan J, Zha X, Chen S, Li Y. Higher TOX Genes Expression Is Associated With Poor Overall Survival for Patients With Acute Myeloid Leukemia. Front Oncol 2021; 11:740642. [PMID: 34692519 PMCID: PMC8532529 DOI: 10.3389/fonc.2021.740642] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Thymocyte selection-associated HMG box (TOX) is a transcription factor that belongs to the high mobility group box (HMG-box) superfamily, which includes four subfamily members: TOX, TOX2, TOX3, and TOX4. TOX is related to the formation of multiple malignancies and contributes to CD8+ T cell exhaustion in solid tumors. However, little is known about the role of TOX genes in hematological malignancies. In this study, we explored the prognostic value of TOX genes from 40 patients with de novo acute myeloid leukemia (AML) by quantitative real-time PCR (qRT-PCR) in a training cohort and validated the results using transcriptome data from 167 de novo AML patients from the Cancer Genome Atlas (TCGA) database. In the training cohort, higher expression of TOX and TOX4 was detected in the AML samples, whereas lower TOX3 expression was found. Moreover, both the training and validation results indicated that higher TOX2, TOX3, and TOX4 expression of AML patients (3-year OS: 0% vs. 37%, P = 0.036; 3-year OS: 4% vs. 61%, P < 0.001; 3-year OS: 0% vs. 32%, P = 0.010) and the AML patients with highly co-expressed TOX, TOX2, TOX4 genes (3-year OS: 0% vs. 25% vs. 75%, P = 0.001) were associated with poor overall survival (OS). Interestingly, TOX2 was positively correlated with CTLA-4, PD-1, TIGIT, and PDL-2 (rs = 0.43, P = 0.006; rs = 0.43, P = 0.006; rs = 0.56, P < 0.001; rs = 0.54, P < 0.001). In conclusion, higher expression of TOX genes was associated with poor OS for AML patients, which was related to the up-regulation of immune checkpoint genes. These data might provide novel predictors for AML outcome and direction for further investigation of the possibility of using TOX genes in novel targeted therapies for AML.
Collapse
Affiliation(s)
- Chaofeng Liang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Yujie Zhao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Cunte Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Shuxin Huang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Tairan Deng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Xiangbo Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Jiaxiong Tan
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Shaohua Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
26
|
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
|
27
|
Wang C, Sun C, Li M, Xia B, Wang Y, Zhang L, Zhang Y, Wang J, Sun F, Lu S, Zhu J, Huang J, Zhang Y. Novel fully human anti-CD47 antibodies stimulate phagocytosis and promote elimination of AML cells. J Cell Physiol 2021; 236:4470-4481. [PMID: 33206395 DOI: 10.1002/jcp.30163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 01/03/2023]
Abstract
Although most patients with acute myeloid leukemia (AML) enter remission after induction chemotherapy, the risk of relapse remains considerable. Therefore, some novel therapeutic strategies are still required. This study found that the overexpression of CD47 on AML cells was at least twofold more than that on normal bone marrow (NBM) cells in 81% (17/21) of the investigated patients; no patients had lower expression level of CD47 compared with healthy donors. The study also demonstrated that blocking the CD47/SIRPα (signal regulatory protein α) signal with the established novel fully human anti-CD47 monoclonal antibodies increased the phagocytosis of AML cells by macrophages in vitro. Furthermore, in vivo experiments showed that the novel fully human anti-CD47 monoclonal antibodies could significantly prolong the survival time of mice. Overall, the novel fully human anti-CD47 antibodies could block CD47/SIRPα interaction, increase macrophage-mediated phagocytosis, and enhance the elimination of AML cells.
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody Specificity
- Antigens, Differentiation/metabolism
- Antineoplastic Agents, Immunological/pharmacology
- Binding Sites, Antibody
- CD47 Antigen/antagonists & inhibitors
- CD47 Antigen/immunology
- CD47 Antigen/metabolism
- Case-Control Studies
- Female
- HL-60 Cells
- Humans
- K562 Cells
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Middle Aged
- Phagocytosis/drug effects
- Receptors, Immunologic/metabolism
- THP-1 Cells
- U937 Cells
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Chaoyu Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Chengtao Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Mengzhen Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bing Xia
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yi Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yanyan Zhang
- INSERM Unité Mixte de Recherche (UMR), Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Juan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Feifei Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Suying Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jia Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Junting Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yizhuo Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Department of Hematology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
28
|
Lee E, Koh Y, Hong J, Eom HS, Yoon SS. Recent Clinical Update of Acute Myeloid Leukemia: Focus on Epigenetic Therapies. J Korean Med Sci 2021; 36:e85. [PMID: 33821592 PMCID: PMC8021975 DOI: 10.3346/jkms.2021.36.e85] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 01/20/2021] [Indexed: 01/05/2023] Open
Abstract
Acute myeloid leukemia (AML) is a complicated disease characterized by genetic heterogeneity and simultaneous alterations in multiple genes. For decades, its only curative method has been intensive induction chemotherapy with or without allogeneic hematopoietic stem cell transplantation, and this approach cannot be applied to elderly patients, who make up more than 50% of AML patients. Recent advances in genomics facilitated the elucidation of various mutations related to AML, and the most frequent mutations were discovered in epigenetic regulators. Alterations to epigenetic modifications that are essential for normal cell biology, including DNA methylation and histone acetylation, have been identified. As epigenetic dysregulation is an important carcinogenic mechanism and some epigenetic changes are reversible, these epigenetic alterations have become targets for novel drug development against AML. This review summarizes the recent advances in epigenetic therapies for AML and discusses future research directions.
Collapse
Affiliation(s)
- Eunyoung Lee
- Department of Internal Medicine, Center for Hematologic Malignancy, National Cancer Center, Goyang, Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Junshik Hong
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyeon Seok Eom
- Department of Internal Medicine, Center for Hematologic Malignancy, National Cancer Center, Goyang, Korea
| | - Sung Soo Yoon
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
29
|
Lyu T, Wang Y, Li D, Yang H, Qin B, Zhang W, Li Z, Cheng C, Zhang B, Guo R, Song Y. Exosomes from BM-MSCs promote acute myeloid leukemia cell proliferation, invasion and chemoresistance via upregulation of S100A4. Exp Hematol Oncol 2021; 10:24. [PMID: 33789743 PMCID: PMC8011411 DOI: 10.1186/s40164-021-00220-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023] Open
Abstract
Background BM-MSCs play an important role in cancer development through the release of cytokines or exosomes. Studies have shown that extracellular exosomes derived from BM-MSCs are a key pro-invasive factor. However, how BM-MSC-exos influence AML cell proliferation, invasion and chemoresistance remains poorly understood. Methods We isolated exosomes from BM-MSCs and used electron microscopy, particle size separation and western blots to identify the exosomes. The invasion of leukemia cells was observed with a transwell assay. The stemness traits and chemoresistance of the leukemia cells were detected by FCM, colony formation and CCK-8 assays. TCGA database was used to investigate the prognostic relevance of S100A4 and its potential role in AML. Results In this study, we found that BM-MSC-exos increased the metastatic potential, maintained the stemness and contributed to the chemoresistance of leukemia cells. Mechanistically, BM-MSC-exos promoted the proliferation, invasion and chemoresistance of leukemia cells via upregulation of S100A4. Downregulating S100A4 clearly suppressed the proliferation, invasion, and chemoresistance of leukemia cells after treatment with BM-MSC-exos. Bioinformatic analysis with data in TCGA database showed that S100A4 was associated with poor prognosis in AML patients, and functional enrichment revealed its involvement in the processes of cell–cell adhesion and cytokine regulation. Conclusions S100A4 is vital in the BM-MSC-exo-driven proliferation, invasion and chemoresistance of leukemia cells and may serve as a potential target for leukemia therapy.
Collapse
Affiliation(s)
- Tianxin Lyu
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Yinuo Wang
- Translational Cancer Research Center, Peking University First Hospital, Beijing, 100034, China
| | - Ding Li
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Hui Yang
- Translational Cancer Research Center, Peking University First Hospital, Beijing, 100034, China
| | - Bin Qin
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Wenli Zhang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Zhiyue Li
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Cheng Cheng
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Binglei Zhang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China.
| |
Collapse
|
30
|
Hansen JD, Correa M, Alexander M, Nagy M, Huang D, Sapienza J, Lu G, LeBrun LA, Cathers BE, Zhang W, Tang Y, Ammirante M, Narla RK, Piccotti JR, Pourdehnad M, Lopez-Girona A. CC-90009: A Cereblon E3 Ligase Modulating Drug That Promotes Selective Degradation of GSPT1 for the Treatment of Acute Myeloid Leukemia. J Med Chem 2021; 64:1835-1843. [PMID: 33591756 DOI: 10.1021/acs.jmedchem.0c01489] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemia (AML) is marked by significant unmet clinical need due to both poor survival and high relapse rates where long-term disease control for most patients with relapsed or refractory AML remain dismal. Inspired to bring novel therapeutic options to these patients, we envisioned protein degradation as a potential therapeutic approach for the treatment of AML. Following this course, we discovered and pioneered a novel mechanism of action which culminated in the discovery of CC-90009. CC-90009 represents a novel protein degrader and the first cereblon E3 ligase modulating drug to enter clinical development that specifically targets GSPT1 (G1 to S phase transition 1) for proteasomal degradation. This manuscript briefly summarizes the mechanism of action, scientific rationale, medicinal chemistry, pharmacokinetic properties, and efficacy data for CC-90009, which is currently in phase 1 clinical development.
Collapse
Affiliation(s)
- Joshua D Hansen
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Matthew Correa
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Matt Alexander
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Mark Nagy
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Dehua Huang
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - John Sapienza
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Gang Lu
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Laurie A LeBrun
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Brian E Cathers
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Weihong Zhang
- Bristol Myers Squibb, 556 Morris Avenue, Summit, New Jersey 07901, United States
| | - Yang Tang
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Massimo Ammirante
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Rama K Narla
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Joseph R Piccotti
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| | - Michael Pourdehnad
- Bristol Myers Squibb, 1500 Owens Street, Suite 600, San Francisco, California 94158, United States
| | - Antonia Lopez-Girona
- Bristol Myers Squibb, 10300 Campus Point Drive, Suite 100, San Diego, California 92121, United States
| |
Collapse
|
31
|
Wang T, Pine AR, Kotini AG, Yuan H, Zamparo L, Starczynowski DT, Leslie C, Papapetrou EP. Sequential CRISPR gene editing in human iPSCs charts the clonal evolution of myeloid leukemia and identifies early disease targets. Cell Stem Cell 2021; 28:1074-1089.e7. [PMID: 33571445 DOI: 10.1016/j.stem.2021.01.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/20/2020] [Accepted: 01/14/2021] [Indexed: 12/20/2022]
Abstract
Human cancers arise through the sequential acquisition of somatic mutations that create successive clonal populations. Human cancer evolution models could help illuminate this process and inform therapeutic intervention at an early disease stage, but their creation has faced significant challenges. Here, we combined induced pluripotent stem cell (iPSC) and CRISPR-Cas9 technologies to develop a model of the clonal evolution of acute myeloid leukemia (AML). Through the stepwise introduction of three driver mutations, we generated iPSC lines that, upon hematopoietic differentiation, capture distinct premalignant stages, including clonal hematopoiesis (CH) and myelodysplastic syndrome (MDS), culminating in a transplantable leukemia, and recapitulate transcriptional and chromatin accessibility signatures of primary human MDS and AML. By mapping dynamic changes in transcriptomes and chromatin landscapes, we characterize transcriptional programs driving specific transitions between disease stages. We identify cell-autonomous dysregulation of inflammatory signaling as an early and persistent event in leukemogenesis and a promising early therapeutic target.
Collapse
Affiliation(s)
- Tiansu Wang
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allison R Pine
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andriana G Kotini
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Han Yuan
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lee Zamparo
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel T Starczynowski
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christina Leslie
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
32
|
Sheng H, Zhang J, Ma Y, Zhang Y, Dai Y, Jiang R. lncRNA FBXL19-AS1 is a diagnosis biomarker for paediatric patients with acute myeloid leukemia. J Gene Med 2021; 23:e3317. [PMID: 33474753 DOI: 10.1002/jgm.3317] [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: 06/05/2020] [Revised: 08/15/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNA) have emerged as novel clinical biomarkers and therapeutic targets for various tumors because of their disease- and stage-restricted expression. lncRNA FBXL19 antisense RNA 1 (FBXL19-AS1) expression has been confirmed to be up-regulated in several tumors. However, its expression and effects in paediatric acute myeloid leukemia (AML) have not been elucidated. METHODS Serum FBXL19-AS1 expression was determined in 137 AML patients compared to 43 healthy controls ( < 0.01). RESULTS Using receiver operating characteristic curve analysis, we observed that serum FBXL19-AS1 provided the highly diagnostic performance for the detection of AML (AUC = 0.841, < 0.001). We also examined the association between serum FBXL19-AS1 expression and clinicopathological factors, finding that its high expression was associated with French-American-British classification ( = 0.011) and cytogenetics ( = 0.021). Survival assays with the Kaplan-Meier method revealed that the overall survival ( = 0.0088) and disease-free-survival ( = 0.0027) of AML patients with high serum FBXL19-AS1 levels were distinctly shorter compared to those with low serum FBXL19-AS1 levels. More importantly, Multivariate analysis identified serum FBXL19-AS1 overexpression as an independent unfavorable prognostic factor for both overall survival and disease-free-survival of AML patients. CONCLUSIONS Overall, our findings revealed that high expression of serum FBXL19-AS1 might be useful as a novel prognostic and diagnostic biomarker for AML patients.
Collapse
Affiliation(s)
- Hongling Sheng
- Pediatric Department, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jiajia Zhang
- Obstetrics and Gynecology Department, Shandong Provincial Third Hospital, Cheeloo College of Medicine, China
| | - Yan Ma
- Pediatric Department, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuhua Zhang
- Rehabilitation Department, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yunpeng Dai
- Pediatric Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Renpeng Jiang
- Pediatric Department, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
33
|
Hu X, Wang Y, Gao X, Xu S, Zang L, Xiao Y, Li Z, Hua H, Xu J, Li D. Recent Progress of Oridonin and Its Derivatives for the Treatment of Acute Myelogenous Leukemia. Mini Rev Med Chem 2020; 20:483-497. [PMID: 31660811 DOI: 10.2174/1389557519666191029121809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/13/2019] [Accepted: 09/06/2019] [Indexed: 01/03/2023]
Abstract
First stage human clinical trial (CTR20150246) for HAO472, the L-alanine-(14-oridonin) ester trifluoroacetate, was conducted by a Chinese company, Hengrui Medicine Co. Ltd, to develop a new treatment for acute myelogenous leukemia. Two patents, WO2015180549A1 and CN201410047904.X, covered the development of the I-type crystal, stability experiment, conversion rate research, bioavailability experiment, safety assessment, and solubility study. HAO472 hewed out new avenues to explore the therapeutic properties of oridonin derivatives and develop promising treatment of cancer originated from naturally derived drug candidates. Herein, we sought to overview recent progress of the synthetic, physiological, and pharmacological investigations of oridonin and its derivatives, aiming to disclose the therapeutic potentials and broaden the platform for the discovery of new anticancer drugs.
Collapse
Affiliation(s)
- Xu Hu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yan Wang
- Valiant Co. Ltd., 11 Wuzhishan Road, YEDA Yantai, Shandong 264006, China
| | - Xiang Gao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Shengtao Xu
- Department of Medicinal Chemistry and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Linghe Zang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yan Xiao
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Zhanlin Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Huiming Hua
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Jinyi Xu
- Department of Medicinal Chemistry and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Dahong Li
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, and School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
34
|
Roussel X, Daguindau E, Berceanu A, Desbrosses Y, Warda W, Neto da Rocha M, Trad R, Deconinck E, Deschamps M, Ferrand C. Acute Myeloid Leukemia: From Biology to Clinical Practices Through Development and Pre-Clinical Therapeutics. Front Oncol 2020; 10:599933. [PMID: 33363031 PMCID: PMC7757414 DOI: 10.3389/fonc.2020.599933] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Recent studies have provided several insights into acute myeloid leukemia. Studies based on molecular biology have identified eight functional mutations involved in leukemogenesis, including driver and passenger mutations. Insight into Leukemia stem cells (LSCs) and assessment of cell surface markers have enabled characterization of LSCs from hematopoietic stem and progenitor cells. Clonal evolution has been described as having an effect similar to that of microenvironment alterations. Such biological findings have enabled the development of new targeted drugs, including drug inhibitors and monoclonal antibodies with blockage functions. Some recently approved targeted drugs have resulted in new therapeutic strategies that enhance standard intensive chemotherapy regimens as well as supportive care regimens. Besides the progress made in adoptive immunotherapy, since allogenic hematopoietic stem cell transplantation enabled the development of new T-cell transfer therapies, such as chimeric antigen receptor T-cell and transgenic TCR T-cell engineering, new promising strategies that are investigated.
Collapse
Affiliation(s)
- Xavier Roussel
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Etienne Daguindau
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Ana Berceanu
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Yohan Desbrosses
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Walid Warda
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | | | - Rim Trad
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | - Eric Deconinck
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Marina Deschamps
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | - Christophe Ferrand
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
35
|
Valent P, Bauer K, Sadovnik I, Smiljkovic D, Ivanov D, Herrmann H, Filik Y, Eisenwort G, Sperr WR, Rabitsch W. Cell-based and antibody-mediated immunotherapies directed against leukemic stem cells in acute myeloid leukemia: Perspectives and open issues. Stem Cells Transl Med 2020; 9:1331-1343. [PMID: 32657052 PMCID: PMC7581453 DOI: 10.1002/sctm.20-0147] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022] Open
Abstract
Despite new insights in molecular features of leukemic cells and the availability of novel treatment approaches and drugs, acute myeloid leukemia (AML) remains a major clinical challenge. In fact, many patients with AML relapse after standard therapy and eventually die from progressive disease. The basic concept of leukemic stem cells (LSC) has been coined with the goal to decipher clonal architectures in various leukemia-models and to develop curative drug therapies by eliminating LSC. Indeed, during the past few years, various immunotherapies have been tested in AML, and several of these therapies follow the strategy to eliminate relevant leukemic subclones by introducing LSC-targeting antibodies or LSC-targeting immune cells. These therapies include, among others, new generations of LSC-eliminating antibody-constructs, checkpoint-targeting antibodies, bi-specific antibodies, and CAR-T or CAR-NK cell-based strategies. However, responses are often limited and/or transient which may be due to LSC resistance. Indeed, AML LSC exhibit multiple forms of resistance against various drugs and immunotherapies. An additional problems are treatment-induced myelotoxicity and other side effects. The current article provides a short overview of immunological targets expressed on LSC in AML. Moreover, cell-based therapies and immunotherapies tested in AML are discussed. Finally, the article provides an overview about LSC resistance and strategies to overcome resistance.
Collapse
Affiliation(s)
- Peter Valent
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Karin Bauer
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Dubravka Smiljkovic
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
| | - Daniel Ivanov
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
| | - Harald Herrmann
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
- Department of Radiation OncologyMedical University of ViennaViennaAustria
| | - Yüksel Filik
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Gregor Eisenwort
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology and HemostaseologyMedical University of ViennaViennaAustria
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - Werner Rabitsch
- Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
- Department of Internal Medicine I, Stem Cell Transplantation UnitMedical University of ViennaViennaAustria
| |
Collapse
|
36
|
Wu J, Xiao Y, Sun J, Sun H, Chen H, Zhu Y, Fu H, Yu C, E W, Lai S, Ma L, Li J, Fei L, Jiang M, Wang J, Ye F, Wang R, Zhou Z, Zhang G, Zhang T, Ding Q, Wang Z, Hao S, Liu L, Zheng W, He J, Huang W, Wang Y, Xie J, Li T, Cheng T, Han X, Huang H, Guo G. A single-cell survey of cellular hierarchy in acute myeloid leukemia. J Hematol Oncol 2020; 13:128. [PMID: 32977829 PMCID: PMC7517826 DOI: 10.1186/s13045-020-00941-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background Acute myeloid leukemia (AML) is a fatal hematopoietic malignancy and has a prognosis that varies with its genetic complexity. However, there has been no appropriate integrative analysis on the hierarchy of different AML subtypes. Methods Using Microwell-seq, a high-throughput single-cell mRNA sequencing platform, we analyzed the cellular hierarchy of bone marrow samples from 40 patients and 3 healthy donors. We also used single-cell single-molecule real-time (SMRT) sequencing to investigate the clonal heterogeneity of AML cells. Results From the integrative analysis of 191727 AML cells, we established a single-cell AML landscape and identified an AML progenitor cell cluster with novel AML markers. Patients with ribosomal protein high progenitor cells had a low remission rate. We deduced two types of AML with diverse clinical outcomes. We traced mitochondrial mutations in the AML landscape by combining Microwell-seq with SMRT sequencing. We propose the existence of a phenotypic “cancer attractor” that might help to define a common phenotype for AML progenitor cells. Finally, we explored the potential drug targets by making comparisons between the AML landscape and the Human Cell Landscape. Conclusions We identified a key AML progenitor cell cluster. A high ribosomal protein gene level indicates the poor prognosis. We deduced two types of AML and explored the potential drug targets. Our results suggest the existence of a cancer attractor.
Collapse
Affiliation(s)
- Junqing Wu
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Yanyu Xiao
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Jie Sun
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Huiyu Sun
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Haide Chen
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Yuanyuan Zhu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Huarui Fu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chengxuan Yu
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Weigao E
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Shujing Lai
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Lifeng Ma
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Jiaqi Li
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Lijiang Fei
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Mengmeng Jiang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Jingjing Wang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Fang Ye
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Renying Wang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Ziming Zhou
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Guodong Zhang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Tingyue Zhang
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China
| | - Qiong Ding
- Wuhan Biobank Co., LTD, Wuhan, 430075, China
| | - Zou Wang
- Wuhan Biobank Co., LTD, Wuhan, 430075, China
| | - Sheng Hao
- Wuhan Biobank Co., LTD, Wuhan, 430075, China
| | - Lizhen Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Weiyan Zheng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Weijia Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yungui Wang
- Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jin Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
| | - Tiefeng Li
- Institute of Applied Mechanics, Zhejiang University, Hangzhou, 310027, China
| | - Tao Cheng
- Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300000, China.,Alliance for Atlas of Blood Cells, Tianjin, China
| | - Xiaoping Han
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China. .,Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China.
| | - He Huang
- Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China. .,Alliance for Atlas of Blood Cells, Tianjin, China. .,Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China.
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,Stem Cell Institute, Zhejiang University, Hangzhou, 310058, China. .,Alliance for Atlas of Blood Cells, Tianjin, China. .,Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
37
|
Cancilla D, Rettig MP, DiPersio JF. Targeting CXCR4 in AML and ALL. Front Oncol 2020; 10:1672. [PMID: 33014834 PMCID: PMC7499473 DOI: 10.3389/fonc.2020.01672] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/29/2020] [Indexed: 12/13/2022] Open
Abstract
The interaction of acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) blasts with the bone marrow microenvironment regulates self-renewal, growth signaling, as well as chemotherapy resistance. The chemokine receptor, CXC receptor 4 (CXCR4), with its ligand chemokine ligand 12 (CXCL12), plays a key role in the survival and migration of normal and malignant stem cells to the bone marrow. High expression of CXCR4 on AML and ALL blasts has been shown to be a predictor of poor prognosis for these diseases. Several small molecule inhibitors, short peptides, antibodies, and antibody drug conjugates have been developed for the purposes of more effective targeting and killing of malignant cells expressing CXCR4. In this review we will discuss recent results and strategies in targeting CXCR4 with these agents in patients with AML or ALL.
Collapse
Affiliation(s)
| | | | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
38
|
Kim Y, Jeung HK, Cheong JW, Song J, Bae SH, Lee JI, Min YH. All-Trans Retinoic Acid Synergizes with Enasidenib to Induce Differentiation of IDH2-Mutant Acute Myeloid Leukemia Cells. Yonsei Med J 2020; 61:762-773. [PMID: 32882760 PMCID: PMC7471081 DOI: 10.3349/ymj.2020.61.9.762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/10/2020] [Accepted: 07/22/2020] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Pharmacological inhibition of mutant isocitrate dehydrogenase (IDH) reduces R-2-hydroxyglutarate (2-HG) levels and restores cellular differentiation in vivo and in vitro. The IDH2 inhibitor enasidenib (AG-221) has been approved by the FDA as a first-in-class inhibitor for the treatment of relapsed or refractory (R/R) IDH2-mutant acute myeloid leukemia (AML). In this study, the effects of a combination of all-trans retinoic acid (ATRA) and AG-221 on AML cell differentiation was explored, along with the mechanisms employed by IDH2-mutant cells in AML. MATERIALS AND METHODS We treated the human AML cell line, IDH2-mutant-TF-1, and primary human AML cells carrying IDH2 mutation with 30 μM AG-221 and 100 nM ATRA, alone or in combination. RESULTS Combined treatment with AG-221 and ATRA inhibited 2-HG production and resulted in synergistic effects on differentiation among IDH2-mutant AML cells and primary AML cells expressing IDH2 mutation. Combined treatment with AG-221 and ATRA altered autophagic activity. AG-221 and ATRA treatment-induced differentiation of IDH2-mutant AML cells was associated with autophagy induction, without suppressing autophagy flux at maturation and degradation stages. A RAF-1/MEK/ERK pathway was founded to be associated with AG-221 and ATRA-induced differentiation in IDH2-mutant AML cells. IDH-associated changes in histone methylation markers decreased after AG-221 and ATRA combination treatment. CONCLUSION Our preliminary evidence indicates that the addition of ATRA to treatments with IDH2 inhibitor may lead to further improvements or increases in response rates in IDH2-mutant AML patients who do not appear to benefit from treatments with IDH2 inhibitor alone.
Collapse
Affiliation(s)
- Yundeok Kim
- Department of Internal Medicine, Yonsei Wonju College of Medicine, Wonju, Korea
| | - Hoi Kyung Jeung
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - June Won Cheong
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jaewoo Song
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Han Bae
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jong In Lee
- Department of Internal Medicine, Yonsei Wonju College of Medicine, Wonju, Korea
| | - Yoo Hong Min
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
39
|
Yamashita M, Dellorusso PV, Olson OC, Passegué E. Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis. Nat Rev Cancer 2020; 20:365-382. [PMID: 32415283 PMCID: PMC7658795 DOI: 10.1038/s41568-020-0260-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2020] [Indexed: 12/17/2022]
Abstract
Haematopoiesis is governed by haematopoietic stem cells (HSCs) that produce all lineages of blood and immune cells. The maintenance of blood homeostasis requires a dynamic response of HSCs to stress, and dysregulation of these adaptive-response mechanisms underlies the development of myeloid leukaemia. Leukaemogenesis often occurs in a stepwise manner, with genetic and epigenetic changes accumulating in pre-leukaemic HSCs prior to the emergence of leukaemic stem cells (LSCs) and the development of acute myeloid leukaemia. Clinical data have revealed the existence of age-related clonal haematopoiesis, or the asymptomatic clonal expansion of mutated blood cells in the elderly, and this phenomenon is connected to susceptibility to leukaemic transformation. Here we describe how selection for specific mutations that increase HSC competitive fitness, in conjunction with additional endogenous and environmental changes, drives leukaemic transformation. We review the ways in which LSCs take advantage of normal HSC properties to promote survival and expansion, thus underlying disease recurrence and resistance to conventional therapies, and we detail our current understanding of leukaemic 'stemness' regulation. Overall, we link the cellular and molecular mechanisms regulating HSC behaviour with the functional dysregulation of these mechanisms in myeloid leukaemia and discuss opportunities for targeting LSC-specific mechanisms for the prevention or cure of malignant diseases.
Collapse
Affiliation(s)
- Masayuki Yamashita
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Paul V Dellorusso
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Oakley C Olson
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
40
|
Chen T, Zhang J, Zeng H, Zhang Y, Zhang Y, Zhou X, Zhou H. Antiproliferative effects of L-asparaginase in acute myeloid leukemia. Exp Ther Med 2020; 20:2070-2078. [PMID: 32782519 PMCID: PMC7401243 DOI: 10.3892/etm.2020.8904] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/06/2019] [Indexed: 01/13/2023] Open
Abstract
The antitumor enzyme L-asparaginase (L-Asp) has commonly been used for the treatment of acute lymphoblastic leukemia. However, the effects of L-Asp on acute myeloid leukemia (AML) and their underlying mechanisms have not been fully elucidated. In the present study, the effects of L-Asp on cell proliferation and apoptosis were investigated using the AML cell lines U937, HL-60 and KG-1a. The effects of combining L-Asp with mitoxantrone (MIT) and cytarabine (Ara-c) were also analyzed. The combination of MIT and Ara-C is known as MA therapy, and is a widely used therapeutic regimen for the treatment of elderly patients with refractory AML. When applied alone, L-Asp inhibited cell proliferation and induced apoptosis in each of the cell lines tested. Furthermore, the combined use of L-Asp with MA therapy further potentiated the inhibition of cell proliferation while increasing the induction of apoptosis. These findings provide evidence for the potential antitumor effect of L-Asp in AML, and indicate that improved efficacy maybe achieved by combining L-Asp with MIT and Ara-c. This combination may provide a promising new therapeutic strategy for the treatment of AML.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Juan Zhang
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Hui Zeng
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Yue Zhang
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Yong Zhang
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Xiaohuan Zhou
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| | - Hebing Zhou
- Department of Hematology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, P.R. China
| |
Collapse
|
41
|
Shifting therapeutic paradigms in induction and consolidation for older adults with acute myeloid leukemia. Curr Opin Hematol 2020; 26:51-57. [PMID: 30585895 DOI: 10.1097/moh.0000000000000480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE OF REVIEW As the age and life expectancy of the general population rise, the number of acute myeloid leukemia (AML) patients suitable for therapy is expected to dramatically increase. The population of older adults with AML, while already comprising the vast majority of AML patients, has not been specifically addressed in terms of unique age-related features, such as existence of comorbidities, frailty, and disease biology. RECENT FINDINGS Over the past decade, major improvements in the approach to the management of older adults with AML included: incorporation of new comorbidity scores specifically oriented to this patient population that can predict individual fitness to treatment, refined knowledge of the unique mutational landscape, and incorporating new combinations and novel agents designed to target the AML biology. Particularly, the recent exciting description of age-related clonal hematopoiesis and its evolution to AML may open new avenues for intervention prior to development of full-blown leukemia. SUMMARY The rising awareness of the unique biology and special needs of older adults with AML has resulted in the design of new studies aiming to target the aberrant mutations and clinical characteristics in this patient population.
Collapse
|
42
|
Frisch A, Rowe JM, Ofran Y. How we treat older patients with acute myeloid leukaemia. Br J Haematol 2020; 191:682-691. [PMID: 32352169 DOI: 10.1111/bjh.16701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
After decades when intensive chemotherapy remained the only effective anti-acute myeloid leukaemia (AML) treatment, a torrent of novel, less toxic agents are about to revolutionise AML therapy. Prolonged remissions with good quality of life become achievable for many patients previously considered only for palliative care because they could not tolerate intensive therapy. As treatment options multiply, the importance of genetic profile is recognised, even for advanced-age patients for whom cure is unlikely. With lack of randomised comparative trials for most treatment regimens, one can only extrapolate data from existing studies to make evidence-based decisions. We herein present seven common clinical scenarios illustrating the complexity of treating older AML patients and describe our approach to their management. In each case, up-to-date data on relevant agents to be offered to a particular patient are discussed. The current review is limited to the drugs, available and approved in the Western world and many promising agents, still under investigation, are not discussed.
Collapse
Affiliation(s)
- Avraham Frisch
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Jacob M Rowe
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Yishai Ofran
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
43
|
Putyrski M, Vakhrusheva O, Bonn F, Guntur S, Vorobyov A, Brandts C, Dikic I, Ernst A. Disrupting the LC3 Interaction Region (LIR) Binding of Selective Autophagy Receptors Sensitizes AML Cell Lines to Cytarabine. Front Cell Dev Biol 2020; 8:208. [PMID: 32296703 PMCID: PMC7137635 DOI: 10.3389/fcell.2020.00208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
Short linear motifs (SLiMs) located in disordered regions of multidomain proteins are important for the organization of protein-protein interaction networks. By dynamic association with their binding partners, SLiMs enable assembly of multiprotein complexes, pivotal for the regulation of various aspects of cell biology in higher organisms. Despite their importance, there is a paucity of molecular tools to study SLiMs of endogenous proteins in live cells. LC3 interacting regions (LIRs), being quintessential for orchestrating diverse stages of autophagy, are a prominent example of SLiMs and mediate binding to the ubiquitin-like LC3/GABARAP family of proteins. The role of LIRs ranges from the posttranslational processing of their binding partners at early stages of autophagy to the binding of selective autophagy receptors (SARs) to the autophagosome. In order to generate tools to study LIRs in cells, we engineered high affinity binders of LIR motifs of three archetypical SARs: OPTN, p62, and NDP52. In an array of in vitro and cellular assays, the engineered binders were shown to have greatly improved affinity and specificity when compared with the endogenous LC3/GABARAP family of proteins, thus providing a unique possibility for modulating LIR interactions in living systems. We exploited these novel tools to study the impact of LIR inhibition on the fitness and the responsiveness to cytarabine treatment of THP-1 cells - a model for studying acute myeloid leukemia (AML). Our results demonstrate that inhibition of LIR of a single autophagy receptor is insufficient to sensitize the cells to cytarabine, while simultaneous inhibition of three LIR motifs in three distinct SARs reduces the IC50 of the chemotherapeutic.
Collapse
Affiliation(s)
- Mateusz Putyrski
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Frankfurt, Germany
| | - Olesya Vakhrusheva
- Department of Medicine, Hematology/Oncology, Goethe-University, Frankfurt, Germany
| | - Florian Bonn
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Suchithra Guntur
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany
| | - Andrew Vorobyov
- Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Frankfurt, Germany
| | - Christian Brandts
- Department of Medicine, Hematology/Oncology, Goethe-University, Frankfurt, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany.,University Cancer Center Frankfurt, Goethe-University, Frankfurt, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences, Frankfurt, Germany
| | - Andreas Ernst
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Frankfurt, Germany
| |
Collapse
|
44
|
Seo B, Kim J, Kim S, Lee E. Bibliometric analysis of studies about acute myeloid leukemia conducted globally from 1999 to 2018. Blood Res 2020; 55:1-9. [PMID: 32269969 PMCID: PMC7106114 DOI: 10.5045/br.2020.55.1.1] [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: 11/08/2019] [Revised: 01/20/2020] [Accepted: 02/03/2020] [Indexed: 01/12/2023] Open
Abstract
A bibliometric study is performed to analyze publication patterns in a specific research area and to establish a landscape model that can be used to quantitatively weigh publications. This study aimed to investigate AML research networks and to conduct a trend-related keyword analysis. We analyzed 48,202 studies about AML published from 1999 to 2019 in the Web of Science Core Collection. The network analysis was conducted using the R&R studio software. The journal Blood had the highest number of published articles with an h-index of 410. The USA had the highest number of total publications (18,719, 38.3%) and research funded by the government, institutions, and pharmaceutical companies (5,436, 10.8%). The institute with the largest number of publications was the MD Anderson Cancer Center. Kantarjian H, Garcia-Manero G, and Ravandi F were the leading authors of publications about AML. Keyword analysis revealed that FLT 3, micro-RNA, and NK cell topics were the hotspots in the cell and gene area in all publications. The overall AML research landscape is popular in the field of translational research as it can identify molecular, cell, and gene studies conducted by different funding agencies, countries, institutions, and author networks. With active funding and support from the Chinese government, the productivity of scientific research is increasing not only in the AML field but also in the medical/health-related science field.
Collapse
Affiliation(s)
- Beomjun Seo
- Department of Public Health, Graduate School, Korea University, Seoul, Korea
| | - Jeeyoon Kim
- Department of Clinical Pharmacy, Graduate School, Cha University, Seoul, Korea
| | - Seungwook Kim
- Graduate School of Interdisciplinary Management, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Eunil Lee
- Department of Public Health, Graduate School, Korea University, Seoul, Korea
| |
Collapse
|
45
|
Epigenetic therapies in acute myeloid leukemia: where to from here? Blood 2020; 134:1891-1901. [PMID: 31697822 DOI: 10.1182/blood.2019003262] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/06/2019] [Indexed: 12/11/2022] Open
Abstract
A hallmark of acute myeloid leukemia (AML) is epigenetic dysregulation, which is initiated by recurrent translocations and/or mutations in transcription factors and chromatin regulators. This manifests as a block in myeloid differentiation and an increase in malignant self-renewal. These common features of AML have led to widespread optimism that epigenetic therapies would dramatically change the natural history of this disease. Although preclinical studies with these drugs fueled this optimism, results from early clinical trials have offered a more sobering message. Here, we provide an overview of epigenetic therapies that are currently approved by therapeutic regulatory authorities across the world and those undergoing early-phase clinical trials. We also discuss the conceptual and molecular factors that may explain some of the disparity between the bench and bedside, as well as emerging avenues for combining the current generation of epigenetic therapies with other classes of agents and the development of novel epigenetic therapies. With further research and development of this exciting class of drugs, we may finally be able to dramatically improve outcomes for patients afflicted with this aggressive and often incurable malignancy.
Collapse
|
46
|
Rubio-Jurado B, Sosa-Quintero LS, Carrasco-Martinez IL, Norato-Delgado A, Garcia-Luna E, Guzmán-Silahua S, Riebeling-Navarro C, Nava-Zavala AH. New biomarkers in non-Hodgkin lymphoma and acute leukemias. Adv Clin Chem 2020; 96:19-53. [PMID: 32362319 DOI: 10.1016/bs.acc.2019.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Biomarkers play a critical role in the medical care of patients with cancer, including in early detection of the disease, diagnostic accuracy, risk stratification, treatment, and follow-up. Biomarkers in hematological malignancies can support the redefinition of the diagnosis and adjustments in the treatment plan. Biomarkers can be classified into 4 categories: (1) protein antigens, (2) cytogenetic abnormalities, (3) genetic polymorphisms, and (4) gene expression. Efforts in genomics, proteomics, and metabolomics to observe new biomarkers that contribute to the development of clinical medicine with greater precision in the strategies that improve prevention, diagnosis, and treatment of patients with malignant hematological disease. New biomarkers should accomplish several issues such as the biological plausibility, methodology used, analytical validation, intellectual property registry, and legal framework of application. This knowledge should be transferred to health professionals who can carry out the process of its implementation in clinical practice.
Collapse
Affiliation(s)
- Benjamín Rubio-Jurado
- Departamento Clínico de Hematologia, Division Onco-Hematologia, UMAE, Hospital de Especialidades (HE), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico; Unidad de Investigación Biomédica 02, UMAE HE, CMNO, IMSS, Guadalajara, Jalisco, Mexico; Extensión, Consulting and Research Division, Universidad de Monterrey, San Pedro Garza Garcia, Mexico
| | - Lluvia Sugey Sosa-Quintero
- Departamento Clínico de Hematologia, Division Onco-Hematologia, UMAE, Hospital de Especialidades (HE), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Ivette Lenina Carrasco-Martinez
- Departamento Clínico de Hematologia, Division Onco-Hematologia, UMAE, Hospital de Especialidades (HE), Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Armando Norato-Delgado
- Servicio de Hematologia, HGZ No. 21, IMSS, Cerro de Picachos 852, Col Jardines oriente, Tepatitlán, Jalisco, Mexico
| | - Eduardo Garcia-Luna
- Vice-Rector, División de Ciencias de la Salud, Universidad de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | - Sandra Guzmán-Silahua
- Unidad de Investigación Biomédica 02, UMAE HE, CMNO, IMSS, Guadalajara, Jalisco, Mexico
| | - Carlos Riebeling-Navarro
- Unidad de Investigación en Epidemiología Clínica, UMAE, Hospital de Pediatría CMNS-XXI, IMSS/UNAM, Mexico City, Mexico
| | - Arnulfo Hernan Nava-Zavala
- Unidad de Investigación Biomédica 02, UMAE HE, CMNO, IMSS, Guadalajara, Jalisco, Mexico; Programa Internacional, Facultad de Medicina, Universidad Autónoma de Guadalajara, Zapopan, Jalisco, Mexico; Departamento de Inmunologia y Reumatologia, Hospital General de Occidente, Secretaria de Salud Jalisco, Zapopan, Jalisco, Mexico.
| |
Collapse
|
47
|
Zhang X, Xu Y, Wang J, Zhao S, Li J, Huang X, Xu H, Zhang X, Suo S, Lv Y, Zhang Y, Yu W. miR-221-3p Delivered by BMMSC-Derived Microvesicles Promotes the Development of Acute Myelocytic Leukemia. Front Bioeng Biotechnol 2020; 8:81. [PMID: 32117949 PMCID: PMC7033425 DOI: 10.3389/fbioe.2020.00081] [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: 12/20/2019] [Accepted: 01/29/2020] [Indexed: 12/20/2022] Open
Abstract
Objective: The study aims to investigate the effects of miR-221-3p in bone marrow mesenchymal stem cell (BMMSC)-derived microvesicles (MVs) on cell cycle, proliferation and invasion of acute myelocytic leukemia (AML). Methods: Bioinformatics was used to predict differentially expressed miRNAs (DEmiRNAs) in AML. The morphology of BMMSC-derived MVs was observed under an electron microscope, and the positional relation of MVs and OCI-AML2 cells was observed by a fluorescence microscope. MTT, Transwell, and flow cytometry assays were used to analyze the effects of MVs on OCI-AML2 cells. The targeted relationship between miR-221-3p and CDKN1C was detected by dual luciferase assay. Results: It was verified that miR-221-3p promoted the proliferation, invasion and migration of OCI-AML2 cells, and induced the cell cycle arrest in G1/S phase as well as inhibited cell apoptosis. Further studies showed that MVs promoted the proliferation, migration and invasion of AML, and induced the cell cycle arrest in G1/S phase through miR-221-3p. It was confirmed that miR-221-3p can directly target CDKN1C to regulate cell cycle, proliferation and invasion of AML. Conclusion: miR-221-3p in BMMSC-derived MVs regulated AML cell cycle, cell proliferation and invasion through targeting CDKN1C. miR-221-3p and CDKN1C were considered to be potential targets and biomarkers for the treatment of AML in clinic.
Collapse
Affiliation(s)
- Xuewu Zhang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yu Xu
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Jinghan Wang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Shuqi Zhao
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Jianhu Li
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Xin Huang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Huan Xu
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Xiang Zhang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Shanshan Suo
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yunfei Lv
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Yi Zhang
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| | - Wenjuan Yu
- Department of Hematology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
| |
Collapse
|
48
|
Nanocarriers as Magic Bullets in the Treatment of Leukemia. NANOMATERIALS 2020; 10:nano10020276. [PMID: 32041219 PMCID: PMC7075174 DOI: 10.3390/nano10020276] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/21/2022]
Abstract
Leukemia is a type of hematopoietic stem/progenitor cell malignancy characterized by the accumulation of immature cells in the blood and bone marrow. Treatment strategies mainly rely on the administration of chemotherapeutic agents, which, unfortunately, are known for their high toxicity and side effects. The concept of targeted therapy as magic bullet was introduced by Paul Erlich about 100 years ago, to inspire new therapies able to tackle the disadvantages of chemotherapeutic agents. Currently, nanoparticles are considered viable options in the treatment of different types of cancer, including leukemia. The main advantages associated with the use of these nanocarriers summarized as follows: i) they may be designed to target leukemic cells selectively; ii) they invariably enhance bioavailability and blood circulation half-life; iii) their mode of action is expected to reduce side effects. FDA approval of many nanocarriers for treatment of relapsed or refractory leukemia and the desired results extend their application in clinics. In the present review, different types of nanocarriers, their capability in targeting leukemic cells, and the latest preclinical and clinical data are discussed.
Collapse
|
49
|
Elgamal OA, Mehmood A, Jeon JY, Carmichael B, Lehman A, Orwick SJ, Truxall J, Goettl VM, Wasmuth R, Tran M, Mitchell S, Lapalombella R, Eathiraj S, Schwartz B, Stegmaier K, Baker SD, Hertlein E, Byrd JC. Preclinical efficacy for a novel tyrosine kinase inhibitor, ArQule 531 against acute myeloid leukemia. J Hematol Oncol 2020; 13:8. [PMID: 31992353 PMCID: PMC6988309 DOI: 10.1186/s13045-019-0821-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is the most common type of adult leukemia. Several studies have demonstrated that oncogenesis in AML is enhanced by kinase signaling pathways such as Src family kinases (SFK) including Src and Lyn, spleen tyrosine kinase (SYK), and bruton's tyrosine kinase (BTK). Recently, the multi-kinase inhibitor ArQule 531 (ARQ 531) has demonstrated potent inhibition of SFK and BTK that translated to improved pre-clinical in vivo activity as compared with the irreversible BTK inhibitor ibrutinib in chronic lymphocytic leukemia (CLL) models. Given the superior activity of ARQ 531 in CLL, and recognition that this molecule has a broad kinase inhibition profile, we pursued its application in pre-clinical models of AML. METHODS The potency of ARQ 531 was examined in vitro using FLT3 wild type and mutated (ITD) AML cell lines and primary samples. The modulation of pro-survival kinases following ARQ 531 treatment was determined using AML cell lines. The effect of SYK expression on ARQ 531 potency was evaluated using a SYK overexpressing cell line (Ba/F3 murine cells) constitutively expressing FLT3-ITD. Finally, the in vivo activity of ARQ 531 was evaluated using MOLM-13 disseminated xenograft model. RESULTS Our data demonstrate that ARQ 531 treatment has anti-proliferative activity in vitro and impairs colony formation in AML cell lines and primary AML cells independent of the presence of a FLT3 ITD mutation. We demonstrate decreased phosphorylation of oncogenic kinases targeted by ARQ 531, including SFK (Tyr416), BTK, and fms-related tyrosine kinase 3 (FLT3), ultimately leading to changes in down-stream targets including SYK, STAT5a, and ERK1/2. Based upon in vitro drug synergy data, we examined ARQ 531 in the MOLM-13 AML xenograft model alone and in combination with venetoclax. Despite ARQ 531 having a less favorable pharmacokinetics profile in rodents, we demonstrate modest single agent in vivo activity and synergy with venetoclax. CONCLUSIONS Our data support consideration of the application of ARQ 531 in combination trials for AML targeting higher drug concentrations in vivo.
Collapse
Affiliation(s)
- Ola A Elgamal
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Abeera Mehmood
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Jae Yoon Jeon
- Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA
| | - Bridget Carmichael
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Amy Lehman
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Shelley J Orwick
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Jean Truxall
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Virginia M Goettl
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Ronni Wasmuth
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Minh Tran
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Shaneice Mitchell
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA
| | | | | | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston Children's Hospital, Boston, MA, USA
| | - Sharyn D Baker
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA.,Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA
| | - Erin Hertlein
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA.
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, 455 Wiseman Hall, 400 West 12th Avenue, Columbus, OH, 43210, USA. .,Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
50
|
Vegi NM, Chakrabortty S, Zegota MM, Kuan SL, Stumper A, Rawat VPS, Sieste S, Buske C, Rau S, Weil T, Feuring-Buske M. Somatostatin receptor mediated targeting of acute myeloid leukemia by photodynamic metal complexes for light induced apoptosis. Sci Rep 2020; 10:371. [PMID: 31941913 PMCID: PMC6962389 DOI: 10.1038/s41598-019-57172-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 12/21/2019] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is characterized by relapse and treatment resistance in a major fraction of patients, underlining the need of innovative AML targeting therapies. Here we analysed the therapeutic potential of an innovative biohybrid consisting of the tumor-associated peptide somatostatin and the photosensitizer ruthenium in AML cell lines and primary AML patient samples. Selective toxicity was analyzed by using CD34 enriched cord blood cells as control. Treatment of OCI AML3, HL60 and THP1 resulted in a 92, and 99 and 97% decrease in clonogenic growth compared to the controls. Primary AML cells demonstrated a major response with a 74 to 99% reduction in clonogenicity in 5 of 6 patient samples. In contrast, treatment of CD34+ CB cells resulted in substantially less reduction in colony numbers. Subcellular localization assays of RU-SST in OCI-AML3 cells confirmed strong co-localization of RU-SST in the lysosomes compared to the other cellular organelles. Our data demonstrate that conjugation of a Ruthenium complex with somatostatin is efficiently eradicating LSC candidates of patients with AML. This indicates that receptor mediated lysosomal accumulation of photodynamic metal complexes is a highly attractive approach for targeting AML cells.
Collapse
Affiliation(s)
- Naidu M Vegi
- Institute of Experimental Cancer Research, Comprehensive Cancer Centre, University Hospital Ulm, D-89081, Ulm, Germany
| | - Sabyasachi Chakrabortty
- Department of Chemistry, SRM University, AP - Amaravati, Andhra Pradesh, 522502, India.,Max Planck Institute for Polymer Research, D-55128, Mainz, Germany
| | - Maksymilian M Zegota
- Max Planck Institute for Polymer Research, D-55128, Mainz, Germany.,Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Seah Ling Kuan
- Max Planck Institute for Polymer Research, D-55128, Mainz, Germany.,Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Anne Stumper
- Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Vijay P S Rawat
- Institute of Experimental Cancer Research, Comprehensive Cancer Centre, University Hospital Ulm, D-89081, Ulm, Germany
| | - Stefanie Sieste
- Max Planck Institute for Polymer Research, D-55128, Mainz, Germany.,Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Christian Buske
- Institute of Experimental Cancer Research, Comprehensive Cancer Centre, University Hospital Ulm, D-89081, Ulm, Germany
| | - Sven Rau
- Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research, D-55128, Mainz, Germany.,Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Michaela Feuring-Buske
- Institute of Experimental Cancer Research, Comprehensive Cancer Centre, University Hospital Ulm, D-89081, Ulm, Germany. .,Department of Internal Medicine III, University Hospital Ulm, D-89081, Ulm, Germany.
| |
Collapse
|