1
|
Kumar ARK, Low J, Lim J, Myint B, Sun X, Wu L, Cheng HS, Yip S, Ming Cheng CZ, Manoharan T, Quek YJ, Shou Y, Tian JS, Ng YY, Gascoigne NRJ, Tan NS, Sugimura R, Chia G, Sze Cheung AM, Yawata M, Tay A. Non-viral, high throughput genetic engineering of primary immune cells using nanostraw-mediated transfection. Biomaterials 2025; 317:123079. [PMID: 39842078 DOI: 10.1016/j.biomaterials.2024.123079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Transfection of proteins, mRNA, and chimeric antigen receptor (CAR) transgenes into immune cells remains a critical bottleneck in cell manufacturing. Current methods, such as viruses and bulk electroporation, are hampered by low transfection efficiency, unintended transgene integration, and significant cell perturbation. The Nanostraw Electro-actuated Transfection (NExT) technology offers a solution by using high aspect-ratio nanostraws and localized electric fields to precisely deliver biomolecules into cells with minimal disruption. We demonstrate that NExT can deliver proteins, polysaccharides, and mRNA into primary human CD8+ and CD4+ T cells, and achieve CRISPR/Cas9 gene knockout of CXCR4 and TRAC in CD8+ T cells. We showcase NExT's versatility across a range of primary human immune cells, including CD4+ T cells, γδ-T cells, dendritic cells, NK cells, Treg cells, macrophages, and neutrophils. Finally, we developed a scalable, high-throughput multiwell NExT system capable of transfecting over 14 million cells and delivering diverse cargoes into multiple cell types from various donors simultaneously. This technology holds promise for streamlining high-throughput screening of allogeneic donors and reducing optimization costs for large-scale CAR-immune cell transfection.
Collapse
Affiliation(s)
- Arun R K Kumar
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Jessalyn Low
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Jet Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Ba Myint
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Xinhong Sun
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Ling Wu
- Immunology Translational Research Programme and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117545, Singapore
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Sophronia Yip
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China; Centre for Translational Stem Cell Biology, Hong Kong SAR, China
| | - Cyrus Zai Ming Cheng
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Thamizhanban Manoharan
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Ying Jie Quek
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Johann Shane Tian
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Yu Yang Ng
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme and Department of Microbiology and Immunology, National University of Singapore, Singapore, 117545, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Rio Sugimura
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China; Centre for Translational Stem Cell Biology, Hong Kong SAR, China
| | - Gloryn Chia
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Alice Man Sze Cheung
- Department of Haematology, Singapore General Hospital, Singapore, 169608, Singapore; SingHealth Duke-NUS Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, 168753, Singapore
| | - Makoto Yawata
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 119077, Singapore
| | - Andy Tay
- Institute of Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore, 117576, Singapore; NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore.
| |
Collapse
|
2
|
Cao J, Hu B, Li T, Fang D, Jiang L, Wang J. Cellular heterogeneity and cytokine signatures in acute myeloid leukemia: A novel prognostic model. Transl Oncol 2025; 52:102194. [PMID: 39689517 PMCID: PMC11719339 DOI: 10.1016/j.tranon.2024.102194] [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: 08/17/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 12/19/2024] Open
Abstract
Acute Myeloid Leukemia (AML) is a complex hematological malignancy distinguished by its heterogeneity in genetic aberrations, cellular composition, and clinical outcomes. This diversity complicates the development of effective, universally applicable therapeutic strategies and highlights the necessity for personalized approaches to treatment. In our study, we utilized high-resolution single-cell RNA sequencing from publicly available datasets to dissect the complex cellular landscape of AML. This approach uncovered a diverse array of cellular subpopulations within the bone marrow samples of AML patients. Through meticulous analysis, we identified 156 differentially expressed cytokine-related genes that underscore the nuanced interplay between AML cells and their microenvironment. Leveraging this comprehensive dataset, we constructed a prognostic risk score model based on seven pivotal cytokine-related genes: CCL23, IL2RA, IL3RA, IL6R, INHBA, TNFSF15, and TNFSF18. The mRNA levels of 7 genes in the risk score model have significant different. This model was rigorously validated across several independent AML patient cohorts, showcasing its robust prognostic capability to stratify patients into distinct risk categories. Patients classified under the high-risk category exhibited significantly poorer survival outcomes compared to their low-risk counterparts, underscoring the model's clinical relevance. Additionally, our in-depth investigation into the immune landscape revealed marked differences in immune cell infiltration and cytokine signaling between the identified risk groups, shedding light on potential immune-mediated mechanisms driving disease progression and treatment resistance. This comprehensive analysis not only advances our understanding of the cellular and molecular underpinnings of AML but also introduces a novel, clinically applicable risk score model. This tool holds significant promise for enhancing the precision of prognostic assessments in AML, thereby paving the way for more tailored and effective therapeutic interventions. Our findings represent a pivotal step toward the realization of personalized medicine in the management of AML, offering new avenues for research and treatment optimization in this challenging disease landscape.
Collapse
Affiliation(s)
- Jinxia Cao
- Department of Hematology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Wuling District, Changde, Hunan Province, China
| | - Bin Hu
- Department of Hematology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Wuling District, Changde, Hunan Province, China
| | - Tianqi Li
- Department of Hematology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Wuling District, Changde, Hunan Province, China
| | - Dan Fang
- Department of Hematology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Wuling District, Changde, Hunan Province, China
| | - Ling Jiang
- Department of Hematology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Wuling District, Changde, Hunan Province, China
| | - Jun Wang
- Department of Hematology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Wuling District, Changde, Hunan Province, China.
| |
Collapse
|
3
|
Xu Y, Li SC, Xiao J, Liu Q, Cherukuri D, Liu Y, Mirshahidi S, Xu J, Chen X, Homa D, Olea J, Wu K, Kelly KR, Sun F, Huang R, Wang X, Wen Q, Zhang X, Ghiuzeli CM, Chong E, Abdel-Azim H, Reeves ME, Baylink DJ, Cao H, Zhong JF. Exploring treatment-driven subclonal evolution of prognostic triple biomarkers: Dual gene fusions and chimeric RNA variants in novel subtypes of acute myeloid leukemia patients with KMT2A rearrangement. Drug Resist Updat 2025; 79:101199. [PMID: 39823827 DOI: 10.1016/j.drup.2024.101199] [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: 08/02/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/20/2025]
Abstract
Chromosomal rearrangements (CR) initiate leukemogenesis in approximately 50 % of acute myeloid leukemia (AML) patients; however, limited targeted therapies exist due to a lack of accurate molecular and genetic biomarkers of refractory mechanisms during treatment. Here, we investigated the pathological landscape of treatment resistance and relapse in 16 CR-AML patients by monitoring cytogenetic, RNAseq, and genome-wide changes among newly diagnosed, refractory, and relapsed AML. First, in FISH-diagnosed KMT2A (MLL gene, 11q23)/AFDN (AF6, 6q27)-rearrangement, RNA-sequencing identified an unknown CCDC32 (15q15.1)/CBX3 (7p15.2) gene fusion in both newly diagnosed and relapsed samples, which is previously unknown in KMT2A/AFDN-rearranged AML patients. Second, the unreported CCDC32/CBX3 gene fusion significantly affected the expression of wild-type genes of both CCDC32 (essential for embryonic development) and CBX3 (an oncogene for solid tumors) during the relapse, as demonstrated by Quantitative PCR analyses. Third, we further confirmed the existence of triple biomarkers - KMT2A/AFDN (AF6, 6q27) rearrangement, the unknown CCDC32 (15q15.1)/CBX3 (7p15.2) gene fusion and chimeric RNA variants (treatment-resistant leukemic blasts harboring distinct breakpoints) in a 21-year-old male patient of rapid relapsed/refractory AML. Most intriguingly, in this work regarding 16 patients, patients 7 and 20 initially showed the KMT2A/AFDN gene fusion; upon relapse, patient 20 did not show this fusion. On the other hand, patient 7 retained the KMT2A/AFDN fusion at diagnosis and during the relapse, only identified by PCR and Sanger's Sequencing, not by cytogenetics. Interestingly, the chimeric CCDC32/CBX3 gene fusion persisted in the 21-year-old male patient over the diagnostic and relapse phases. Most intriguingly, the overexpression of CCDC32/CBX3 fusion gene in AML patient-specific MV4-11 cells confirms the functional validation, providing experimental evidence of the biological impact of the CCDC32/CBX3 fusion on AML pathogenesis and treatment resistance by promoting cell cycle progression, a mechanism through which AML evolves to become treatment-resistant. All these might exhort differential resistance to treatment. Thus, we found that prognostic and predictive triple biomarkers - KRAS mutated, dual fusions (KMT2A/AFDN, CCDC32/CBX3), and chimeric variants - might evolve with a potential oncogenic role of subclonal evolution for poor clinical outcomes.
Collapse
Affiliation(s)
- Yi Xu
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States; Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States.
| | - Shengwen Calvin Li
- Department of Neurology, University of California-Irvine School of Medicine, 200 S. Manchester Ave. Ste. 206, Orange, CA 92868, United States; CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC®), Rady Children's Health, 1201 La Veta Ave, Orange, CA 92868-3874, United States.
| | - Jeffrey Xiao
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Qian Liu
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S Maryland Pkwy, Las Vegas, NV 89154, United States; School of Life Sciences, College of Sciences, University of Nevada, 4505 S Maryland Pkwy, Las Vegas, NV 89154, United States
| | - Durga Cherukuri
- Department of Pathology & Laboratory Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Yan Liu
- Department of Pathology & Laboratory Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Saied Mirshahidi
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jane Xu
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, United States
| | - Xuelian Chen
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Dadrastoussi Homa
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Julian Olea
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Kaijin Wu
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Kevin R Kelly
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Fengzhu Sun
- Quantitative and Computational Biology Department, University of Southern California, Los Angeles, CA, United States
| | - Ruihao Huang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Qin Wen
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Cristina M Ghiuzeli
- Division of Hematology, University of Washington School of Medicine, Seattle, WA, United States
| | - Esther Chong
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Hisham Abdel-Azim
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, United States; Division of Transplant and Cell Therapy/Hematological Malignancies, Departments of Pediatrics, Loma Linda University, Loma Linda, CA 92354, United States
| | - Mark E Reeves
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Huynh Cao
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Jiang F Zhong
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, United States.
| |
Collapse
|
4
|
Kantarjian HM, DiNardo CD, Kadia TM, Daver NG, Altman JK, Stein EM, Jabbour E, Schiffer CA, Lang A, Ravandi F. Acute myeloid leukemia management and research in 2025. CA Cancer J Clin 2025; 75:46-67. [PMID: 39656142 PMCID: PMC11745214 DOI: 10.3322/caac.21873] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 01/30/2025] Open
Abstract
The first 5 decades of research in acute myeloid leukemia (AML) were dominated by the cytarabine plus anthracyclines backbone, with advances in strategies including allogeneic hematopoietic stem cell transplantation, high-dose cytarabine, supportive care measures, and targeted therapies for the subset of patients with acute promyelocytic leukemia. Since 2017, a turning point in AML research, 12 agents have received regulatory approval for AML in the United States: venetoclax (BCL2 inhibitor); gemtuzumab ozogamicin (CD33 antibody-drug conjugate); midostaurin, gilteritinib, and quizartinib (fms-like tyrosine kinase 3 inhibitors); ivosidenib, olutasidenib, and enasidenib (isocitrate dehydrogenase 1 and 2 inhibitors); oral azacitidine (a partially absorbable formulation); CPX351 (liposomal encapsulation of cytarabine:daunorubicin at a molar ratio of 5:1); glasdegib (hedgehog inhibitor); and recently revumenib (menin inhibitor; approved November 2024). Oral decitabine-cedazuridine, which is approved as a bioequivalent alternative to parenteral hypomethylating agents in myelodysplastic syndrome, can be used for the same purpose in AML. Menin inhibitors, CD123 antibody-drug conjugates, and other antibodies targeting CD123, CD33, and other surface markers are showing promising results. Herein, the authors review the frontline and later line therapies in AML and discuss important research directions.
Collapse
Affiliation(s)
- Hagop M. Kantarjian
- Department of LeukemiaThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Courtney D. DiNardo
- Department of LeukemiaThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Tapan M. Kadia
- Department of LeukemiaThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Naval G. Daver
- Department of LeukemiaThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Jessica K. Altman
- Division of Hematology/OncologyDepartment of MedicineRobert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityChicagoIllinoisUSA
| | - Eytan M. Stein
- Leukemia ServiceDepartment of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Elias Jabbour
- Department of LeukemiaThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Charles A. Schiffer
- Karmanos Cancer CenterWayne State University School of MedicineDetroitMichiganUSA
| | - Amy Lang
- START Center for Cancer CareSan AntonioTexasUSA
| | - Farhad Ravandi
- Department of LeukemiaThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
5
|
Lu J, Bhakta P, Hwang H, Lachowiez C, Apostolidou E. Clinical Outcomes of Patients With Newly Diagnosed Acute Myeloid Leukemia Receiving Treatment in a Safety-Net Hospital System. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024:S2152-2650(24)02447-9. [PMID: 39799045 DOI: 10.1016/j.clml.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND 'Standard of care' therapies for adult acute myeloid leukemia (AML) have yielded 5-year overall survival (OS) rates of 30%-45 %. Risk stratification and novel targeted therapies have improved 5-year OS rates to >75 % for certain groups in specialized centers. PATIENTS AND METHODS This is a retrospective cohort analysis of outcomes in patients ≥18 years with newly diagnosed AML treated between 2005 and 2019 in the Harris Health County, Safety-Net Hospital System in Houston, TX. RESULTS 192 patients were identified. Median age was 52 years, 52 % were male and 57 % identified as Hispanic. Most patients were uninsured or indigent, receiving care under the county's financial assistance programs (62 %). Of the 184 response-assessable patients, 139 achieved composite complete remission (CRc) (76 %). 182 patients had indications for HCT and only 25 patients received HCT (14 %), with main reasons including noncitizenship status and financial/insurance constraints. The 5-year OS rate in the entire cohort was 30 % (35 % in patients <60 years and 16 % if ≥60 years), with 92 % of deaths attributed to AML-related complications. Early death (<4 weeks) rate was 2 %. Secondary, adverse-risk AML, and uninsured status all portended significantly worse OS rates, per multivariate analysis. Patients with indications for HCT who received this modality fared significantly better than those who did not receive it (5-year OS 54 % vs. 21 %). CONCLUSIONS Optimizing AML remission induction regimens, reducing medication costs, ensuring timely administration of AML directed therapies, enhancing equity and diversity in clinical trials, and addressing socioeconomic factors may improve leukemia care for underserved patients.
Collapse
Affiliation(s)
- Jason Lu
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX
| | - Preeya Bhakta
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX
| | - Hyunsoo Hwang
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX
| | - Curtis Lachowiez
- Division of Hematology/Medical Oncology, Oregon Health and Science University, Portland, OR
| | - Effrosyni Apostolidou
- Section of Benign Hematology, Department of Internal Medicine, MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
6
|
Vinod Kumar K, Kumar A, Kundal K, Sengupta A, R K, Singh S, Korra BT, Sharma S, Suresh V, Nishana M, Kumar R. AMLdb: a comprehensive multi-omics platform to identify biomarkers and drug targets for acute myeloid leukemia. Brief Funct Genomics 2024; 23:798-805. [PMID: 38864430 DOI: 10.1093/bfgp/elae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/14/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Acute myeloid leukemia (AML) is one of the leading leukemic malignancies in adults. The heterogeneity of the disease makes the diagnosis and treatment extremely difficult. With the advent of next-generation sequencing (NGS) technologies, exploration at the molecular level for the identification of biomarkers and drug targets has been the focus for the researchers to come up with novel therapies for better prognosis and survival outcomes of AML patients. However, the huge amount of data from NGS platforms requires a comprehensive AML platform to streamline literature mining efforts and save time. To facilitate this, we developed AMLdb, an interactive multi-omics platform that allows users to query, visualize, retrieve, and analyse AML related multi-omics data. AMLdb contains 86 datasets for gene expression profiles, 15 datasets for methylation profiles, CRISPR-Cas9 knockout screens of 26 AML cell lines, sensitivity of 26 AML cell lines to 288 drugs, mutations in 41 unique genes in 23 AML cell lines, and information on 41 experimentally validated biomarkers. In this study, we have reported five genes, i.e. CBFB, ENO1, IMPDH2, SEPHS2, and MYH9 identified via our analysis using AMLdb. ENO1 is uniquely identified gene which requires further investigation as a novel potential target while other reported genes have been previously confirmed as targets through experimental studies. Top of form we believe that these findings utilizing AMLdb can make it an invaluable resource to accelerate the development of effective therapies for AML and assisting the research community in advancing their understanding of AML pathogenesis. AMLdb is freely available at https://project.iith.ac.in/cgntlab/amldb.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Molecular Targeted Therapy
- Genomics/methods
- Databases, Genetic
- Multiomics
Collapse
Affiliation(s)
- Keerthana Vinod Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Ambuj Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Kavita Kundal
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Avik Sengupta
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Kunjulakshmi R
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
- Department of Biological Sciences, Indian Institute of Science Education and Research, Berhampur, Odisha 760010, India
| | - Subashani Singh
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Bhanu Teja Korra
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Simran Sharma
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Vandana Suresh
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Mayilaadumveettil Nishana
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Maruthamala P. O., Vithura, Kerala 695551, India
| | - Rahul Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| |
Collapse
|
7
|
Ji L, Yang W, Gao LJ, Zhang BY, Xu YQ, Xu XF. Treatment of acute myeloid leukemia with active pulmonary tuberculosis with venetoclax‑based anti‑acute myeloid leukemia regimen combined with an intensive and then individualized anti‑tuberculosis regimen: A report of two cases. Oncol Lett 2024; 28:601. [PMID: 39493431 PMCID: PMC11529375 DOI: 10.3892/ol.2024.14734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 06/11/2024] [Indexed: 11/05/2024] Open
Abstract
Patients with concurrent acute myeloid leukemia (AML) and active pulmonary tuberculosis (TB) exhibit certain characteristics; cough, phlegm, fever, hemoptysis, weight loss and dyspnea are common symptoms of both diseases. These patients often cannot tolerate traditional intensive chemotherapy regimens, and finding the optimal timing in the treatment of both AML and active pulmonary TB is complex. Neglecting timely treatment can lead to serious complications and even fatal outcomes. The present paper reports two cases of patients with AML who were diagnosed with active pulmonary TB. The patients received intensive anti-TB treatment with isoniazid, rifampicin, pyrazinamide and ethambutol for 10-15 days. After three consecutive negative sputum smears, the patients in cases 1 and 2 were treated with a venetoclax, homoharringtonine and cytarabine regimen; and a venetoclax and azacitidine regimen for anti-AML therapy, respectively, as well as individualized anti-TB regimens of isoniazid, pyrazinamide, ethambutol and quinolone. Subsequently, both patients achieved complete remission of AML and their active TB was well controlled.
Collapse
Affiliation(s)
- Lin Ji
- Department of Oncology and Hematology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Wei Yang
- Department of Oncology and Hematology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Liu Jie Gao
- Department of Oncology and Hematology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Bei Yuan Zhang
- Department of Oncology and Hematology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Ya Qing Xu
- Department of Oncology and Hematology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Xiao Feng Xu
- Department of Oncology and Hematology, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
8
|
Bhagwat AS, Torres L, Shestova O, Shestov M, Mellors PW, Fisher HR, Farooki SN, Frost BF, Loken MR, Gaymon AL, Frazee D, Rogal W, Frey N, Hexner EO, Luger SM, Loren AW, Martin ME, McCurdy SR, Perl AE, Stadtmauer EA, Brogdon JL, Fraietta JA, Hwang WT, Siegel DL, Plesa G, Aplenc R, Porter DL, June CH, Gill SI. Cytokine-mediated CAR T therapy resistance in AML. Nat Med 2024; 30:3697-3708. [PMID: 39333315 DOI: 10.1038/s41591-024-03271-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 08/27/2024] [Indexed: 09/29/2024]
Abstract
Acute myeloid leukemia (AML) is a rapidly progressive malignancy without effective therapies for refractory disease. So far, chimeric antigen receptor (CAR) T cell therapy in AML has not recapitulated the efficacy seen in B cell malignancies. Here we report a pilot study of autologous anti-CD123 CAR T cells in 12 adults with relapsed or refractory AML. CAR T cells targeting CD123+ cells were successfully manufactured in 90.4% of runs. Cytokine release syndrome was observed in 10 of 12 infused individuals (83.3%, 90% confidence interval 0.5-0.97). Three individuals achieved clinical response (25%, 90% confidence interval 0.07-0.53). We found that myeloid-supporting cytokines are secreted during cell therapy and support AML blast survival via kinase signaling, leading to CAR T cell exhaustion. The prosurvival effect of therapy-induced cytokines presents a unique resistance mechanism in AML that is distinct from any observed in B cell malignancies. Our findings suggest that autologous CART manufacturing is feasible in AML, but treatment is associated with high rates of cytokine release syndrome and relatively poor clinical efficacy. Combining CAR T cell therapies with cytokine signaling inhibitors could enhance immunotherapy efficacy in AML and achieve improved outcomes (ClinicalTrials.gov identifier: NCT03766126 ).
Collapse
Affiliation(s)
- Anand S Bhagwat
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonel Torres
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Shestova
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maksim Shestov
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick W Mellors
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Han R Fisher
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Saamia N Farooki
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Benjamin F Frost
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Avery L Gaymon
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Diane Frazee
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter Rogal
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Noelle Frey
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth O Hexner
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Selina M Luger
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alison W Loren
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Ellen Martin
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon R McCurdy
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander E Perl
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Stadtmauer
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Joseph A Fraietta
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei-Ting Hwang
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Don L Siegel
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Aplenc
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David L Porter
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Bin T, Tang J, Lu B, Xu XJ, Lin C, Wang Y. Construction of AML prognostic model with CYP2E1 and GALNT12 biomarkers based on golgi- associated genes. Ann Hematol 2024:10.1007/s00277-024-06119-7. [PMID: 39604595 DOI: 10.1007/s00277-024-06119-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Acute myeloid leukaemia (AML) was originally an aggressive malignancy of the bone marrow and one of the deadliest forms of acute leukaemia. The 5-year mortality benefit for patients with AML was only 28.3%. Moreover, a large proportion of patients experienced frequent relapses even after remission, thus predicting a bleak prognosis. This research employed differential expression analysis of AML and normal samples sourced from the GSE30029 database, as well as weighted gene co-expression network analysis (WGCNA). We discovered differential golgi apparatus-related genes (DGARGs) specifically associated with AML. Via regressivity analysis and machine learning algorithm, the cancer genome atlas-acute myeloid leukemia (TCGA-AML) cohort developed a prognostic model using characteristic prognostic genes. The performance value of risk score was analysed using Kaplan-Meier (KM) curves and Cox regression. A predictive nomogram was developed to assess the outcome. The association between prognostic trait genes and the immune microenvironment was examined. Finally, immunoactivity and drug susceptibilities were evaluated in various risk groups identified by prognostic signature genes. A total of 77 DGARGs were obtained by differential expression analysis with WGCNA analysis. Following univariate Cox regression and LASSO regression, six prognostic signature genes (ARL5B, GALNT12, MANSC1, PDE4DIP, NCALD and CYP2E1) were utilized to develop a prognostic model. This model was calibrated via KM survival and receiver operating characteristic (ROC) curves, which concluded that it had a predictive impact on the prognosis of AML. Further analysis of the tumour microenvironment in AML patients demonstrated notable variances in immune cell APC_co_inhibition, CCR, Parainflammation, Type_I_IFN_Response, and Type_II_IFN_Response between the high-risk and low-risk groups. A prognostic model was devised in this study using six prognostic genes linked to the Golgi apparatus. The exactness of the model in guiding the prognosis of AML was established. As a result of expression validation, CYP2E1 and GALNT12 will be used as biomarkers to offer fresh insights into the prognosis and treatment of AML patients.
Collapse
Affiliation(s)
- Ting Bin
- Department of Haematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China
| | - Jing Tang
- Department of Haematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China
| | - Bo Lu
- Department of Haematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China
| | - Xiao-Jun Xu
- Department of Haematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China.
| | - Chao Lin
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China.
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, 410000, China.
| | - Ying Wang
- Department of Haematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China.
| |
Collapse
|
10
|
Yan M, Luo X, Han H, Qiu J, Ye Q, Zhang L, Wang Y. ROCK2 increases drug resistance in acute myeloid leukemia via metabolic reprogramming and MAPK/PI3K/AKT signaling. Int Immunopharmacol 2024; 140:112897. [PMID: 39126734 DOI: 10.1016/j.intimp.2024.112897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/17/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Rho-associated coiled-coil kinase 2 (ROCK2) is classified as a member of the serine/threonine protein kinase family and has been identified as a key driver of the development of various forms of cancer. The cause of ROCK2's impact on acute myeloid leukemia (AML) is still unknown. We found that ROCK2 expression was higher in AML patients, leading to lower complete response rates and worse overall survival. Additionally, ROCK2 expression was elevated in the doxorubicin-resistant leukemia cell line HL-60/ADM when compared to their individual parent cells. Moreover, the suppression or inhibition of ROCK2 leads to enhanced drug sensitivity in both AML cell lines and primary AML specimens, along with a notable decrease in downstream signaling pathways. Furthermore, the suppression of ROCK2 caused disruption of cellular energy production pathways by directly affecting the functionality of proteins within the mitochondrial electron transport chain. Finally, we discovered that TRIM26, a specific E3 ligase, is capable of ubiquitylating ROCK2, and the upregulation of TRIM26 within HL-60/ADM cells resulted in heightened sensitivity to the drug and reduced resistance. Thus, our study presents a new strategy for overcoming drug resistance in AML through targeting ROCK2/AKT/MAPK signaling pathway.
Collapse
Affiliation(s)
- Muxia Yan
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xin Luo
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Hong Han
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Jiachun Qiu
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qian Ye
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Li Zhang
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Yiqian Wang
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Battiwalla M, Chao JH, Cox T, Cruz JC, Donnellan WB, Eghtedar A, Kambhampati S, Malik S, Maris MB, Rotta M, Slovick FT, Ramakrishnan A, Bhushan V, Sears L, Martin C, Holder J, Junglen A, Majhail N, LeMaistre CF. Determinants of Outcomes for Acute Myeloid Leukemia Patients Treated in a Community-Based Specialized Versus Non-Specialized Hospital Setting. Clin Hematol Int 2024; 6:43-52. [PMID: 39417014 PMCID: PMC11477936 DOI: 10.46989/001c.124273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/29/2023] [Indexed: 10/19/2024] Open
Abstract
The treatment setting influences acute myeloid leukemia (AML) outcomes. Most cancer patients receive care in the community, yet few studies have evaluated the effectiveness of clinical programs outside of academic or National Cancer Institute (NCI)-designated cancer centers. This was a multi-level, case-controlled study of real-world outcomes for initial AML treatment in a community-based network for 1,391 patients with AML between 2011 and 2018. We benchmarked survival within our network against the Surveillance, Epidemiology, and End Results (SEER) database. Coarsened exact matching was performed against 17,186 chemotherapy-treated patients in the SEER database. Cox proportional and accelerated failure time multivariable modeling were performed to identify patient, disease, therapy and center characteristics associated with the risk of AML mortality. Within the network, 799 patients were treated at six specialized blood cancer centers and 592 at 63 other hospitals. Patients receiving high-intensity induction at specialized centers had improved median survivals of 31 months versus 18 months [P<0.001] at non-specialized centers. Median survivals were 13 for non-specialized centers versus 10 months for SEER [P<0.001], and 18 for the entire network versus 10 months for SEER [P<0.001]. Multivariable modeling showed significant impacts from age (HR = 1.025), high-intensity induction therapy (HR= .695), diagnosis year (HR= .937), neighborhood income (HR = .997; P<0.01), higher acuity (HR = 1.002) and Charlson comorbidity score (HR = 1.117). AML treatment may be effectively delivered in the community hospital setting, with specialized centers producing better outcomes for higher intensity treatments.
Collapse
Affiliation(s)
- Minoo Battiwalla
- Sarah Cannon Blood Cancer Network
- Sarah Cannon Center for Blood Cancer at TriStar Centennial
| | - Ju-Hsien Chao
- Sarah Cannon Blood Cancer Network
- Texas Transplant Institute
| | | | | | | | - Alireza Eghtedar
- Sarah Cannon Blood Cancer Network
- Colorado Blood Cancer Institute
| | - Suman Kambhampati
- Sarah Cannon Blood Cancer Network
- HCA Midwest Health at Research Medical Center
| | - Shahbaz Malik
- Sarah Cannon Blood Cancer Network
- Texas Transplant Institute
| | - Michael B. Maris
- Sarah Cannon Blood Cancer Network
- Colorado Blood Cancer Institute
| | - Marcello Rotta
- Sarah Cannon Blood Cancer Network
- Colorado Blood Cancer Institute
| | - Frank T. Slovick
- Sarah Cannon Blood Cancer Network
- HCA Midwest Health at Research Medical Center
| | | | - Vikas Bhushan
- Sarah Cannon Blood Cancer Network
- Medical City Dallas Hospital
| | - Lindsay Sears
- Healthcare Corporations of America Research Institute
| | - Casey Martin
- Healthcare Corporations of America Research Institute
| | - Jared Holder
- Healthcare Corporations of America Research Institute
| | | | | | - Charles F. LeMaistre
- Sarah Cannon Blood Cancer Network
- Sarah Cannon Center for Blood Cancer at TriStar Centennial
| |
Collapse
|
12
|
Cao H, Xiao J, Baylink DJ, Nguyen V, Shim N, Lee J, Mallari DJR, Wasnik S, Mirshahidi S, Chen CS, Abdel-Azim H, Reeves ME, Xu Y. Development of a Competitive Nutrient-Based T-Cell Immunotherapy Designed to Block the Adaptive Warburg Effect in Acute Myeloid Leukemia. Biomedicines 2024; 12:2250. [PMID: 39457563 PMCID: PMC11504511 DOI: 10.3390/biomedicines12102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: T-cell-based adoptive cell therapies have emerged at the forefront of cancer immunotherapies; however, failed long-term survival and inevitable exhaustion of transplanted T lymphocytes in vivo limits clinical efficacy. Leukemia blasts possess enhanced glycolysis (Warburg effect), exploiting their microenvironment to deprive nutrients (e.g., glucose) from T cells, leading to T-cell dysfunction and leukemia progression. Methods: Thus, we explored whether genetic reprogramming of T-cell metabolism could improve their survival and empower T cells with a competitive glucose-uptake advantage against blasts and inhibit their uncontrolled proliferation. Results: Here, we discovered that high-glucose concentration reduced the T-cell expression of glucose transporter GLUT1 (SLC2A1) and TFAM (mitochondrion transcription factor A), an essential transcriptional regulator of mitochondrial biogenesis, leading to their impaired expansion ex vivo. To overcome the glucose-induced genetic deficiency in metabolism, we engineered T cells with lentiviral overexpression of SLC2A1 and/or TFAM transgene. Multi-omics analyses revealed that metabolic reprogramming promoted T-cell proliferation by increasing IL-2 release and reducing exhaustion. Moreover, the engineered T cells competitively deprived glucose from allogenic blasts and lessened leukemia burden in vitro. Conclusions: Our findings propose a novel T-cell immunotherapy that utilizes a dual strategy of starving blasts and cytotoxicity for preventing uncontrolled leukemia proliferation.
Collapse
Affiliation(s)
- Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Vinh Nguyen
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Nathan Shim
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jae Lee
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Dave J. R. Mallari
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Saied Mirshahidi
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Biospecimen Laboratory, Department of Medicine and Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hisham Abdel-Azim
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Transplant and Cell Therapy, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
- Division of Hematology and Oncology, Department of Pediatrics, Loma Linda University, Loma Linda, CA 92354, USA
| | - Mark E. Reeves
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yi Xu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
13
|
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
|
14
|
Kantarjian H, Borthakur G, Daver N, DiNardo CD, Issa G, Jabbour E, Kadia T, Sasaki K, Short NJ, Yilmaz M, Ravandi F. Current status and research directions in acute myeloid leukemia. Blood Cancer J 2024; 14:163. [PMID: 39300079 PMCID: PMC11413327 DOI: 10.1038/s41408-024-01143-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
The understanding of the molecular pathobiology of acute myeloid leukemia (AML) has spurred the identification of therapeutic targets and the development of corresponding novel targeted therapies. Since 2017, twelve agents have been approved for the treatment of AML subsets: the BCL2 inhibitor venetoclax; the CD33 antibody drug conjugate gemtuzumab ozogamicin; three FLT3 inhibitors (midostaurin, gilteritinib, quizartinib); three IDH inhibitors (ivosidenib and olutasidenib targeting IDH1 mutations; enasidenib targeting IDH2 mutations); two oral hypomethylating agents (oral poorly absorbable azacitidine; fully absorbable decitabine-cedazuridine [latter approved as an alternative to parenteral hypomethylating agents in myelodysplastic syndrome and chronic myelomonocytic leukemia but commonly used in AML]); and CPX-351 (encapsulated liposomal 5:1 molar ratio of cytarabine and daunorubicin), and glasdegib (hedgehog inhibitor). Other targeted therapies (menin inhibitors, CD123 antibody-drug conjugates) are showing promising results. To achieve optimal results in such a rare and heterogeneous entity as AML requires expertise, familiarity with this rare cancer, and the access to, and delivery of disparate therapies under rigorous supportive care conditions. In this review, we update the standard-of-care and investigational therapies and outline promising current and future research directions.
Collapse
Affiliation(s)
- Hagop Kantarjian
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA.
| | - Gautam Borthakur
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney D DiNardo
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Ghayas Issa
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- From the Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
15
|
McCall D, Abuasab T, Rodriguez-Sevilla JJ, Mohamed SF, Patnaik A, Devireddy K, Arani N, Sheikh I, Jamshidi R, Gibson A, Roth M, Nuñez C, Garcia M, Chien KS, Loghavi S, Pierce SA, Sasaki K, Issa G, Cuglievan B, Kantarjian H, Garcia-Manero G. Characteristics and outcomes of children, adolescent, and young adult patients with myelodysplastic neoplasms: A single-center retrospective analysis. Leuk Res 2024; 144:107563. [PMID: 39178611 DOI: 10.1016/j.leukres.2024.107563] [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: 05/29/2024] [Revised: 08/13/2024] [Accepted: 08/17/2024] [Indexed: 08/26/2024]
Abstract
Myelodysplastic syndrome, or myelodysplastic neoplasms, are a rare finding in pediatric, adolescent, and young adult (AYA) patients. More literature is needed to highlight trends of survival or treatment resistance in subpopulations to improve treatment. Here we report a single center retrospective analysis of pediatric and AYA patients from 2000 to 2022 including molecular and cytogenetic data. Using the IPSS-R and IPSS-M, which have been reported exclusively in adults, and excluding patients with bone marrow failure syndromes, we analyzed 119 pediatric and AYA patients with myelodysplastic neoplasms. Therapy-related myelodysplastic neoplasms were present in 36 % of patients, and 31 % of patients developed acute myeloid leukemia. The 5-year overall survival (OS) rate for the entire cohort was 45 %. Contrary to young adults and older adults, mutations were not common in pediatrics. Those who underwent stem cell transplant (SCT)(at any time) had significantly longer median OS. Although SCT at any time improved OS in the de novo myelodysplastic neoplasm group, the choice of the initial treatment with intensive chemotherapy, hypomethylating agents, or SCT did not significantly alter OS. Median OS was shorter in the pediatric group (<18 years old) and longer for those with isolated deletion of 5q or TET2 mutation, but these were not significant findings. Median OS was significantly shorter in those with monosomy 7 or 7q deletion and those with therapy-related myelodysplastic neoplasms. These findings build on previously reported findings and encourage the use of SCT along with molecular and cytogenetic analysis.
Collapse
Affiliation(s)
- David McCall
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Tareq Abuasab
- Department of Medicine, Baylor University, Houston, TX, USA
| | | | - Shehab Fareed Mohamed
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anish Patnaik
- McGovern Medical School, University of Texas Health Sciences Center at Houston (UTHealth), Houston, TX, USA
| | - Kirthi Devireddy
- McGovern Medical School, University of Texas Health Sciences Center at Houston (UTHealth), Houston, TX, USA
| | - Naszrin Arani
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Irtiza Sheikh
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raehannah Jamshidi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Amber Gibson
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Roth
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cesar Nuñez
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Miriam Garcia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly S Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanam Loghavi
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sherry A Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ghayas Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Branko Cuglievan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
16
|
Salman H. Comparative Analysis of AML Classification Systems: Evaluating the WHO, ICC, and ELN Frameworks and Their Distinctions. Cancers (Basel) 2024; 16:2915. [PMID: 39199685 PMCID: PMC11352995 DOI: 10.3390/cancers16162915] [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: 07/26/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Comprehensive analyses of the molecular heterogeneity of acute myelogenous leukemia, AML, particularly when malignant cells retain normal karyotype, has significantly evolved. In 2022, significant revisions were introduced in the World Health Organization (WHO) classification and the European LeukemiaNet (ELN) 2022 guidelines of acute myeloid leukemia (AML). These revisions coincided with the inception of the first version of the International Consensus Classification (ICC) for AML. These modifications aim to improve diagnosis and treatment outcomes via a comprehensive incorporation of sophisticated genetic and clinical parameters as well as facilitate accruals to innovative clinical trials. Key updates include modifications to the blast count criteria for AML diagnosis, with WHO 2022 eliminating the ≥20% blast requirement in the presence of AML-defining abnormalities and ICC 2022 setting a 10% cutoff for recurrent genetic abnormalities. Additionally, new categories, such as AML with mutated TP53 and MDS/AML, were introduced. ELN 2022 guidelines retained risk stratification approach and emphasized the critical role of measurable residual disease (MRD) that increased the use of next-generation sequencing (NGS) and flow cytometry testing. These revisions underscore the importance of precise classification for targeted treatment strategies and improved patient outcomes. How much difference versus concordance these classifications present and the impact of those on clinical practice is a continuing discussion.
Collapse
Affiliation(s)
- Huda Salman
- Brown Center for Immunotherapy, Melvin and Bren Simon Comprehensive Cancer Center, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
17
|
Xu Y, Baylink DJ, Xiao J, Tran L, Nguyen V, Park B, Valladares I, Lee S, Codorniz K, Tan L, Chen CS, Abdel-Azim H, Reeves ME, Mirshahidi H, Marcucci G, Cao H. Discovery of NFκB2-Coordinated Dual Regulation of Mitochondrial and Nuclear Genomes Leads to an Effective Therapy for Acute Myeloid Leukemia. Int J Mol Sci 2024; 25:8532. [PMID: 39126100 PMCID: PMC11313218 DOI: 10.3390/ijms25158532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Acute myeloid leukemia (AML) has a poor survival rate for both pediatric and adult patients due to its frequent relapse. To elucidate the bioenergetic principle underlying AML relapse, we investigated the transcriptional regulation of mitochondrial-nuclear dual genomes responsible for metabolic plasticity in treatment-resistant blasts. Both the gain and loss of function results demonstrated that NFκB2, a noncanonical transcription factor (TF) of the NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells) family, can control the expression of TFAM (mitochondrial transcription factor A), which is known to be essential for metabolic biogenesis. Furthermore, genetic tracking and promoter assays revealed that NFκB2 is in the mitochondria and can bind the specific "TTGGGGGGTG" region of the regulatory D-loop domain to activate the light-strand promoter (LSP) and heavy-strand promoter 1 (HSP1), promoters of the mitochondrial genome. Based on our discovery of NFκB2's novel function of regulating mitochondrial-nuclear dual genomes, we explored a novel triplet therapy including inhibitors of NFκB2, tyrosine kinase, and mitochondrial ATP synthase that effectively eliminated primary AML blasts with mutations of the FMS-related receptor tyrosine kinase 3 (FLT3) and displayed minimum toxicity to control cells ex vivo. As such, effective treatments for AML must include strong inhibitory actions on the dual genomes mediating metabolic plasticity to improve leukemia prognosis.
Collapse
Affiliation(s)
- Yi Xu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.-S.C.)
- Division Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - David J. Baylink
- Division Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jeffrey Xiao
- Division Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Lily Tran
- Division Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Vinh Nguyen
- Division Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Brandon Park
- Division Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Ismael Valladares
- Division Regenerative Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Scott Lee
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Kevin Codorniz
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Laren Tan
- Division of Pulmonary, Critical Care, Hyperbaric and Sleep Medicine, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.-S.C.)
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hisham Abdel-Azim
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.-S.C.)
- Division of Transplant and Cell Therapy, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
- Division of Hematology and Oncology, Department of Pediatrics, Loma Linda University, Loma Linda, CA 92354, USA
| | - Mark E. Reeves
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.-S.C.)
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hamid Mirshahidi
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.-S.C.)
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Guido Marcucci
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, City of Hope Medical Center and Beckman Research Institute, Duarte, CA 91010, USA
| | - Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (C.-S.C.)
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
18
|
Yuan R, Xu ZJ, Zhang SK, Cao XY, Dai AG, Song L. New evidence for a role of DANCR in cancers: a comprehensive review. J Transl Med 2024; 22:569. [PMID: 38877534 PMCID: PMC11177382 DOI: 10.1186/s12967-024-05246-z] [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: 03/22/2024] [Accepted: 04/28/2024] [Indexed: 06/16/2024] Open
Abstract
Cancer remains a leading cause of mortality and poses a substantial threat to public health. Studies have revealed that Long noncoding RNA DANCR is a cytoplasmic lncRNA whose aberrant expression plays a pivotal role in various cancer types. Within tumour biology, DANCR exerts regulatory control over crucial processes such as proliferation, invasion, metastasis, angiogenesis, inflammatory responses, cellular energy metabolism reprogramming, and apoptosis. By acting as a competitive endogenous RNA for miRNAs and by interacting with proteins and mRNAs at the molecular level, DANCR contributes significantly to cancer progression. Elevated DANCR levels have also been linked to heightened resistance to anticancer drugs. Moreover, the detection of circulating DANCR holds promise as a valuable biomarker for aiding in the clinical differentiation of different cancer types. This article offers a comprehensive review and elucidation of the primary functions and molecular mechanisms through which DANCR influences tumours.
Collapse
Affiliation(s)
- Rong Yuan
- School of Medicine, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China
| | - Zhao-Jun Xu
- Department of Cardiothoracic Surgery, the First Affiliated Hospital, Hunan University of Chinese Medicine, 97 Shaoshan Road, Changsha, 410007, Hunan, China
| | - Sheng-Kang Zhang
- Department of Cardiothoracic Surgery, the First Affiliated Hospital, Hunan University of Chinese Medicine, 97 Shaoshan Road, Changsha, 410007, Hunan, China
| | - Xian-Ya Cao
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Ai-Guo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China.
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China.
| | - Lan Song
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
19
|
Sedeta E, Jemal A, Nisotel L, Sung H. Survival difference between secondary and de novo acute myeloid leukemia by age, antecedent cancer types, and chemotherapy receipt. Cancer 2024; 130:1952-1963. [PMID: 38244208 DOI: 10.1002/cncr.35214] [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: 09/09/2023] [Revised: 11/13/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND This study compared the survival of persons with secondary acute myeloid leukemia (sAML) to those with de novo AML (dnAML) by age at AML diagnosis, chemotherapy receipt, and cancer type preceding sAML diagnosis. METHODS Data from Surveillance, Epidemiology, and End Results 17 Registries were used, which included 47,704 individuals diagnosed with AML between 2001 and 2018. Multivariable Cox proportional hazards regression was used to compare AML-specific survival between sAML and dnAML. Trends in 5-year age-standardized relative survival were examined via the Joinpoint survival model. RESULTS Overall, individuals with sAML had an 8% higher risk of dying from AML (hazard ratio [HR], 1.08; 95% confidence interval [CI], 1.05-1.11) compared to those with dnAML. Disparities widened with younger age at diagnosis, particularly in those who received chemotherapy for AML (HR, 1.14; 95% CI, 1.10-1.19). In persons aged 20-64 years and who received chemotherapy, HRs were greatest for those with antecedent myelodysplastic syndrome (HR, 2.04; 95% CI, 1.83-2.28), ovarian cancer (HR, 1.91; 95% CI, 1.19-3.08), head and neck cancer (HR, 1.55; 95% CI, 1.02-2.36), leukemia (HR, 1.45; 95% CI, 1.12-1.89), and non-Hodgkin lymphoma (HR, 1.42; 95% CI, 1.20-1.69). Among those aged ≥65 years and who received chemotherapy, HRs were highest for those with antecedent cervical cancer (HR, 2.42; 95% CI, 1.15-5.10) and myelodysplastic syndrome (HR, 1.28; 95% CI, 1.19-1.38). The 5-year relative survival improved 0.3% per year for sAML slower than 0.86% per year for dnAML. Consequently, the survival gap widened from 7.2% (95% CI, 5.4%-9.0%) during the period 2001-2003 to 14.3% (95% CI, 12.8%-15.8%) during the period 2012-2014. CONCLUSIONS Significant survival disparities exist between sAML and dnAML on the basis of age at diagnosis, chemotherapy receipt, and antecedent cancer, which highlights opportunities to improve outcomes among those diagnosed with sAML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/epidemiology
- Middle Aged
- Female
- Male
- Adult
- Aged
- SEER Program
- Young Adult
- Age Factors
- Neoplasms, Second Primary/mortality
- Neoplasms, Second Primary/epidemiology
- Aged, 80 and over
- Adolescent
- Proportional Hazards Models
- United States/epidemiology
- Lymphoma, Non-Hodgkin/mortality
- Lymphoma, Non-Hodgkin/drug therapy
- Lymphoma, Non-Hodgkin/epidemiology
- Neoplasms/mortality
- Neoplasms/drug therapy
- Neoplasms/epidemiology
Collapse
Affiliation(s)
- Ephrem Sedeta
- Brookdale University Hospital Medical Center, Brooklyn, New York, USA
| | - Ahmedin Jemal
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| | - Lauren Nisotel
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| | - Hyuna Sung
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| |
Collapse
|
20
|
Jin P, Wang X, Jin Q, Zhang Y, Shen J, Jiang G, Zhu H, Zhao M, Wang D, Li Z, Zhou Y, Li W, Zhang W, Liu Y, Wang S, Jin W, Cao Y, Sheng G, Dong F, Wu S, Li X, Jin Z, He M, Liu X, Chen L, Zhang Y, Wang K, Li J. Mutant U2AF1-Induced Mis-Splicing of mRNA Translation Genes Confers Resistance to Chemotherapy in Acute Myeloid Leukemia. Cancer Res 2024; 84:1583-1596. [PMID: 38417135 DOI: 10.1158/0008-5472.can-23-2543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/07/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
Patients with primary refractory acute myeloid leukemia (AML) have a dismal long-term prognosis. Elucidating the resistance mechanisms to induction chemotherapy could help identify strategies to improve AML patient outcomes. Herein, we retrospectively analyzed the multiomics data of more than 1,500 AML cases and found that patients with spliceosome mutations had a higher risk of developing refractory disease. RNA splicing analysis revealed that the mis-spliced genes in refractory patients converged on translation-associated pathways, promoted mainly by U2AF1 mutations. Integrative analyses of binding and splicing in AML cell lines substantiated that the splicing perturbations of mRNA translation genes originated from both the loss and gain of mutant U2AF1 binding. In particular, the U2AF1S34F and U2AF1Q157R mutants orchestrated the inclusion of exon 11 (encoding a premature termination codon) in the eukaryotic translation initiation factor 4A2 (EIF4A2). This aberrant inclusion led to reduced eIF4A2 protein expression via nonsense-mediated mRNA decay. Consequently, U2AF1 mutations caused a net decrease in global mRNA translation that induced the integrated stress response (ISR) in AML cells, which was confirmed by single-cell RNA sequencing. The induction of ISR enhanced the ability of AML cells to respond and adapt to stress, contributing to chemoresistance. A pharmacologic inhibitor of ISR, ISRIB, sensitized U2AF1 mutant cells to chemotherapy. These findings highlight a resistance mechanism by which U2AF1 mutations drive chemoresistance and provide a therapeutic approach for AML through targeting the ISR pathway. SIGNIFICANCE U2AF1 mutations induce the integrated stress response by disrupting splicing of mRNA translation genes that improves AML cell fitness to enable resistance to chemotherapy, which can be targeted to improve AML treatment.
Collapse
Affiliation(s)
- Peng Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoling Wang
- Department of Reproductive Medical Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiqi Jin
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Yi Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Shen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ge Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongming Zhu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeyi Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zhou
- Department of Reproductive Medical Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenzhu Li
- Department of Reproductive Medical Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yabin Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Siyang Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuncan Cao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guangying Sheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyi Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shishuang Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyang Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengke He
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaxin Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Yunxiang Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kankan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- CNRS-LIA Hematology and Cancer, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junmin Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Wan P, Zhong L, Yu L, Shen C, Shao X, Chen S, Zhou Z, Wang M, Zhang H, Liu B. Lysosome-related genes predict acute myeloid leukemia prognosis and response to immunotherapy. Front Immunol 2024; 15:1384633. [PMID: 38799454 PMCID: PMC11117069 DOI: 10.3389/fimmu.2024.1384633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Background Acute myeloid leukemia (AML) is a highly aggressive and pathogenic hematologic malignancy with consistently high mortality. Lysosomes are organelles involved in cell growth and metabolism that fuse to form specialized Auer rods in AML, and their role in AML has not been elucidated. This study aimed to identify AML subtypes centered on lysosome-related genes and to construct a prognostic model to guide individualized treatment of AML. Methods Gene expression data and clinical data from AML patients were downloaded from two high-throughput sequencing platforms. The 191 lysosomal signature genes were obtained from the database MsigDB. Lysosomal clusters were identified by unsupervised consensus clustering. The differences in molecular expression, biological processes, and the immune microenvironment among lysosomal clusters were subsequently analyzed. Based on the molecular expression differences between lysosomal clusters, lysosomal-related genes affecting AML prognosis were screened by univariate cox regression and multivariate cox regression analyses. Algorithms for LASSO regression analyses were employed to construct prognostic models. The risk factor distribution, KM survival curve, was applied to evaluate the survival distribution of the model. Time-dependent ROC curves, nomograms and calibration curves were used to evaluate the predictive performance of the prognostic models. TIDE scores and drug sensitivity analyses were used to explore the implication of the model for AML treatment. Results Our study identified two lysosomal clusters, cluster1 has longer survival time and stronger immune infiltration compared to cluster2. The differences in biological processes between the two lysosomal clusters are mainly manifested in the lysosomes, vesicles, immune cell function, and apoptosis. The prognostic model consisting of six prognosis-related genes was constructed. The prognostic model showed good predictive performance in all three data sets. Patients in the low-risk group survived significantly longer than those in the high-risk group and had higher immune infiltration and stronger response to immunotherapy. Patients in the high-risk group showed greater sensitivity to cytarabine, imatinib, and bortezomib, but lower sensitivity to ATRA compared to low -risk patients. Conclusion Our prognostic model based on lysosome-related genes can effectively predict the prognosis of AML patients and provide reference evidence for individualized immunotherapy and pharmacological chemotherapy for AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/diagnosis
- Lysosomes/metabolism
- Prognosis
- Female
- Male
- Immunotherapy/methods
- Biomarkers, Tumor/genetics
- Middle Aged
- Gene Expression Profiling
- Adult
- Nomograms
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
- Aged
- Gene Expression Regulation, Leukemic
- Transcriptome
Collapse
Affiliation(s)
- Peng Wan
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Lihua Yu
- Clinical Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Chenlan Shen
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xin Shao
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Shuyu Chen
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Ziwei Zhou
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Meng Wang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Hongyan Zhang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Murtadha M, Park M, Zhu Y, Caserta E, Napolitano O, Tandoh T, Moloudizargari M, Pozhitkov A, Singer M, Dona AA, Vahed H, Gonzalez A, Ly K, Ouyang C, Sanchez JF, Nigam L, Duplan A, Chowdhury A, Ghoda L, Li L, Zhang B, Krishnan A, Marcucci G, Williams JC, Pichiorri F. A CD38-directed, single-chain T-cell engager targets leukemia stem cells through IFN-γ-induced CD38 expression. Blood 2024; 143:1599-1615. [PMID: 38394668 PMCID: PMC11103097 DOI: 10.1182/blood.2023021570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 02/25/2024] Open
Abstract
ABSTRACT Treatment resistance of leukemia stem cells (LSCs) and suppression of the autologous immune system represent major challenges to achieve a cure in acute myeloid leukemia (AML). Although AML blasts generally retain high levels of surface CD38 (CD38pos), LSCs are frequently enriched in the CD34posCD38neg blast fraction. Here, we report that interferon gamma (IFN-γ) reduces LSCs clonogenic activity and induces CD38 upregulation in both CD38pos and CD38neg LSC-enriched blasts. IFN-γ-induced CD38 upregulation depends on interferon regulatory factor 1 transcriptional activation of the CD38 promoter. To leverage this observation, we created a novel compact, single-chain CD38-CD3 T-cell engager (BN-CD38) designed to promote an effective immunological synapse between CD38pos AML cells and both CD8pos and CD4pos T cells. We demonstrate that BN-CD38 engages autologous CD4pos and CD8pos T cells and CD38pos AML blasts, leading to T-cell activation and expansion and to the elimination of leukemia cells in an autologous setting. Importantly, BN-CD38 engagement induces the release of high levels of IFN-γ, driving the expression of CD38 on CD34posCD38neg LSC-enriched blasts and their subsequent elimination. Critically, although BN-CD38 showed significant in vivo efficacy across multiple disseminated AML cell lines and patient-derived xenograft models, it did not affect normal hematopoietic stem cell clonogenicity and the development of multilineage human immune cells in CD34pos humanized mice. Taken together, this study provides important insights to target and eliminate AML LSCs.
Collapse
Affiliation(s)
- Mariam Murtadha
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Miso Park
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Yinghui Zhu
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
- Research Center for Translational Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Enrico Caserta
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ottavio Napolitano
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Theophilus Tandoh
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Milad Moloudizargari
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Alex Pozhitkov
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Mahmoud Singer
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ada Alice Dona
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Hawa Vahed
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Asaul Gonzalez
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Kevin Ly
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ching Ouyang
- Integrative Genomics Core, City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA
| | - James F. Sanchez
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
| | - Lokesh Nigam
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Amanda Duplan
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Arnab Chowdhury
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Lucy Ghoda
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ling Li
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Bin Zhang
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
| | - Guido Marcucci
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - John C. Williams
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Flavia Pichiorri
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| |
Collapse
|
23
|
Entezari M, Tayari A, Paskeh MDA, Kheirabad SK, Naeemi S, Taheriazam A, Dehghani H, Salimimoghadam S, Hashemi M, Mirzaei S, Samarghandian S. Curcumin in treatment of hematological cancers: Promises and challenges. J Tradit Complement Med 2024; 14:121-134. [PMID: 38481552 PMCID: PMC10927384 DOI: 10.1016/j.jtcme.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/16/2023] [Accepted: 10/19/2023] [Indexed: 11/01/2024] Open
Abstract
Hematological cancers include leukemia, myeloma and lymphoma and up to 178.000 new cases are diagnosed with these tumors each year. Different kinds of treatment including radiotherapy, chemotherapy, immunotherapy and stem cell transplantation have been employed in the therapy of hematological cancers. However, they are still causing death among patients. On the other hand, curcumin as an anti-cancer agent for the suppression of human cancers has been introduced. The treatment of hematological cancers using curcumin has been followed. Curcumin diminishes viability and survival rate of leukemia, myeloma and lymphoma cells. Curcumin stimulates apoptosis and G2/M arrest to impair progression of tumor. Curcumin decreases levels of matrix metalloproteinases in suppressing cancer metastasis. A number of downstream targets including VEGF, Akt and STAT3 undergo suppression by curcumin in suppressing progression of hematological cancers. Curcumin stimulates DNA damage and reduces resistance of cancer cells to irradiation. Furthermore, curcumin causes drug sensitivity of hematological tumors, especially myeloma. For targeted delivery of curcumin and improving its pharmacokinetic and anti-cancer features, nanostructures containing curcumin and other anti-cancer agents have been developed.
Collapse
Affiliation(s)
- Maliheh Entezari
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Armita Tayari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Simin Khorsand Kheirabad
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sahar Naeemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Dehghani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Medical Laboratory Sciences, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
24
|
Lim JJ, Othus M, Shaw CM, Russell K, Halpern AB, Appelbaum JS, Hendrie P, Walter RB, Estey EH, Percival MEM. Time independent factors that predict relapse in adults with acute myeloid leukemia. Blood Cancer J 2024; 14:5. [PMID: 38221523 PMCID: PMC10788333 DOI: 10.1038/s41408-023-00954-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 01/16/2024] Open
Affiliation(s)
- John J Lim
- University of Washington Medical Center, Seattle, WA, USA
| | - Megan Othus
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Carole M Shaw
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kathryn Russell
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Anna B Halpern
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jacob S Appelbaum
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Paul Hendrie
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Elihu H Estey
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mary-Elizabeth M Percival
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
25
|
Sljivic I, Fulford A, Ho J, Lazo-Langner A, Xenocostas A, Deotare U. Outpatient consolidation chemotherapy with intermediate dose cytarabine has similar survival and relapses rates in acute myeloid leukemia as compared to high dose cytarabine: A single center analysis. Eur J Haematol 2023; 111:888-894. [PMID: 37640495 DOI: 10.1111/ejh.14094] [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: 05/11/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION The last decade has seen advances in delivering outpatient consolidation therapy for acute myeloid leukemia (AML). The standard of care involves high-dose cytarabine or intermediate-dose cytarabine, given twice daily for three alternating days. At the London Regional Cancer Program, we have transitioned the administration of outpatient cytarabine to a once-daily regimen over six consecutive days. The outcomes of a longer duration interval of high-dose cytarabine and intermediate-dose cytarabine is currently unknown. This study aims to assess the feasibility of administering a continuous 6-day protocol of high-dose (HDAC-16) and intermediate-dose cytarabine (IDAC-16) consolidation therapy in the outpatient setting. METHODS This is a retrospective chart review to analyze AML patients treated with outpatient high-dose or intermediate-dose cytarabine consolidation therapy at the London Regional Cancer Program from January 1, 2019, through November 1, 2022. The primary objective was to determine the outcomes of the 6-day outpatient administration of once daily high-dose cytarabine or intermediate-dose cytarabine. RESULTS Forty-five patients received 89 cycles of cytarabine as outpatients; males were 55.6% of the total population, with a median age of ~57 years. Our overall 2-year survival of HDAC-16 (57.1%) and IDAC-16 (83.3%) is consistent with the reported literature. There was no difference in delays, relapse rates, and nonrelapse mortality between both HDAC and IDAC groups. The 2-year relapse free survival was 57.1% for HDAC-16 and 66.7% for IDAC-16. CONCLUSION Outpatient administration of intermediate-dose cytarabine once daily over six consecutive days results in similar overall survival and relapse rates as compared to high dose cytarabine consolidation chemotherapy. Moving to a once daily administration schedule can alleviate logistical and/or accessibility hurdles for outpatient oncology clinics. Prospective randomized trials are needed in this setting to validate our results.
Collapse
Affiliation(s)
- Igor Sljivic
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Adrienne Fulford
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Jenny Ho
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
- Division of Hematology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Alejandro Lazo-Langner
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
- Division of Hematology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Anargyros Xenocostas
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
- Division of Hematology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Uday Deotare
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
- Division of Hematology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- The Centre for Quality, Innovation and Safety, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
26
|
Xu Y, Baylink DJ, Chen CS, Tan L, Xiao J, Park B, Valladares I, Reeves ME, Cao H. Transient TKI-resistant CD44+pBAD+ blasts undergo intrinsic homeostatic adaptation to promote the survival of acute myeloid leukemia in vitro. Front Oncol 2023; 13:1286863. [PMID: 38023123 PMCID: PMC10664142 DOI: 10.3389/fonc.2023.1286863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 12/01/2023] Open
Abstract
Acute myeloid leukemia (AML) patients have frequent mutations in FMS-like receptor tyrosine kinase 3 (FLT3-mut AML), who respond poorly to salvage chemotherapies and targeted therapies such as tyrosine kinase inhibitors (TKIs). Disease relapse is a common reason of treatment failures in FLT3-mut AML patients, but its intracellular refractory mechanism remains to be discovered. In this study, we designed serial in vitro time-course studies to investigate the biomarkers of TKI-resistant blasts and their survival mechanism. First, we found that a group of transient TKI-resistant blasts were CD44+Phosphorylated-BAD (pBAD)+ and that they could initiate the regrowth of blast clusters in vitro. Notably, TKI-treatments upregulated the compensation pathways to promote PIM2/3-mediated phosphorylation of BAD to initiate the blast survival. Next, we discovered a novel process of intracellular adaptive responses in these transient TKI-resistant blasts, including upregulated JAK/STAT signaling pathways for PIM2/3 expressions and activated SOCS1/SOCS3/PIAS2 inhibitory pathways to down-regulate redundant signal transduction and kinase phosphorylation to regain intracellular homeostasis. Finally, we found that the combination of TKIs with TYK2/STAT4 pathways-driven inhibitors could effectively treat FLT3-mut AML in vitro. In summary, our findings reveal that TKI-treatment can activate a JAK/STAT-PIM2/3 axis-mediated signaling pathways to promote the survival of CD44+pBAD+blasts in vitro. Disrupting these TKIs-activated redundant pathways and blast homeostasis could be a novel therapeutic strategy to treat FLT3-mut AML and prevent disease relapse in vivo.
Collapse
Affiliation(s)
- Yi Xu
- Division of Hematology and Oncology, Loma Linda University Medical Center and Loma Linda University Cancer Center, Loma Linda University Health, Loma Linda, CA, United States
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda University Health, Loma Linda, CA, United States
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda University Health, Loma Linda, CA, United States
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Loma Linda University Medical Center and Loma Linda University Cancer Center, Loma Linda University Health, Loma Linda, CA, United States
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda University Health, Loma Linda, CA, United States
| | - Laren Tan
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda University Health, Loma Linda, CA, United States
- Department of Pulmonary, Critical Care, Hyperbaric and Sleep Medicine, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Brandon Park
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Ismael Valladares
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Mark E. Reeves
- Division of Hematology and Oncology, Loma Linda University Medical Center and Loma Linda University Cancer Center, Loma Linda University Health, Loma Linda, CA, United States
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda University Health, Loma Linda, CA, United States
| | - Huynh Cao
- Division of Hematology and Oncology, Loma Linda University Medical Center and Loma Linda University Cancer Center, Loma Linda University Health, Loma Linda, CA, United States
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda University Health, Loma Linda, CA, United States
| |
Collapse
|
27
|
Wang N. Analysis of prognostic biomarker models and immune microenvironment in acute myeloid leukemia by integrative bioinformatics. J Cancer Res Clin Oncol 2023; 149:9609-9619. [PMID: 37222809 DOI: 10.1007/s00432-023-04871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological cancer driven on by aberrant myeloid precursor cell proliferation and differentiation. A prognostic model was created in this study to direct therapeutic care. METHODS Differentially expressed genes (DEGs) were investigated using the RNA-seq data from the TCGA-LAML and GTEx. Weighted Gene Coexpression Network Analysis (WGCNA) examines the genes involved in cancer. Find the intersection genes and construct the PPI network to discover hub genes and remove prognosis-related genes. A nomogram was produced for predicting the prognosis of AML patients using the risk prognosis model that was constructed using COX and Lasso regression analysis. GO, KEGG, and ssGSEA analysis were used to look into its biological function. TIDE score predicts immunotherapy response. RESULTS Differentially expressed gene analysis revealed 1004 genes, WGCNA analysis revealed 19,575 tumor-related genes, and 941 intersection genes in total. Twelve prognostic genes were found using the PPI network and prognostic analysis. To build a risk rating model, RPS3A and PSMA2 were examined using COX and Lasso regression analysis. The risk score was used to divide the patients into two groups, and Kaplan-Meier analysis indicated that the two groups had different overall survival rates. Univariate and multivariate COX studies demonstrated that risk score is an independent prognostic factor. According to the TIDE study, the immunotherapy response was better in the low-risk group than in the high-risk group. CONCLUSIONS We eventually selected out two molecules to construct prediction models that might be used as biomarkers for predicting AML immunotherapy and prognosis.
Collapse
Affiliation(s)
- Naihong Wang
- Lanzhou University Second Hospital, Gansu, 730000, China.
| |
Collapse
|
28
|
Hussein BA, Kristenson L, Pesce S, Wöhr A, Tian Y, Hallner A, Brune M, Hellstrand K, Tang KW, Bernson E, Thorén FB. NKG2A gene variant predicts outcome of immunotherapy in AML and modulates the repertoire and function of NK cells. J Immunother Cancer 2023; 11:e007202. [PMID: 37648262 PMCID: PMC10471874 DOI: 10.1136/jitc-2023-007202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND The natural killer (NK) complex (NKC) harbors multiple genes such as KLRC1 (encoding NKG2A) and KLRK1 (encoding NKG2D) that are central to regulation of NK cell function. We aimed at determining to what extent NKC haplotypes impact on NK cell repertoire and function, and whether such gene variants impact on outcome of IL-2-based immunotherapy in acute myeloid leukemia (AML). METHODS Genotype status of NKG2D rs1049174 and NKG2A rs1983526 was determined using the TaqMan-Allelic discrimination approach. To dissect the impact of single nucloetide polymorphim (SNP) on NK cell function, we engineered the K562 cell line with CRISPR to be killed in a highly NKG2D-dependent fashion. NK cells were assayed for degranulation, intracellular cytokine production and cytotoxicity using flow cytometry. RESULTS In AML patients receiving immunotherapy, the NKG2A gene variant, rs1983526, was associated with superior leukemia-free survival and overall survival. We observed that superior NK degranulation from individuals with the high-cytotoxicity NKG2D variant was explained by presence of a larger, highly responsive NKG2A+ subset. Notably, NK cells from donors homozygous for a favorable allele encoding NKG2A mounted stronger cytokine responses when challenged with leukemic cells, and NK cells from AML patients with this genotype displayed higher accumulation of granzyme B during histamine dihydrochloride/IL-2 immunotherapy. Additionally, among AML patients, the NKG2A SNP defined a subset of patients with HLA-B-21 TT with a strikingly favorable outcome. CONCLUSIONS The study results imply that a dimorphism in the NKG2A gene is associated with enhanced NK cell effector function and improved outcome of IL-2-based immunotherapy in AML.
Collapse
Affiliation(s)
- Brwa Ali Hussein
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Linnea Kristenson
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Silvia Pesce
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Dipartimento di Medicina Sperimentale, Università di Genova, Genoa, Italy
| | - Anne Wöhr
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Yarong Tian
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Alexander Hallner
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mats Brune
- Department of Hematology, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Kristoffer Hellstrand
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Ka-Wei Tang
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Elin Bernson
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, University of Gothenburg,Gothenburg, Gothenburg, Sweden
| | - Fredrik B Thorén
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
29
|
Lee I, Doepner M, Weissenrieder J, Majer AD, Mercado S, Estell A, Natale CA, Sung PJ, Foskett JK, Carroll MP, Ridky TW. LNS8801 inhibits Acute Myeloid Leukemia by Inducing the Production of Reactive Oxygen Species and Activating the Endoplasmic Reticulum Stress Pathway. CANCER RESEARCH COMMUNICATIONS 2023; 3:1594-1606. [PMID: 37599786 PMCID: PMC10438922 DOI: 10.1158/2767-9764.crc-22-0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/24/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
Despite recent therapeutic advances, the 5-year survival rate for adults with acute myeloid leukemia (AML) is poor and standard-of-care chemotherapy is associated with significant toxicity, highlighting the need for new therapeutic approaches. Recent work from our group and others established that the G protein-coupled estrogen receptor (GPER) is tumor suppressive in melanoma and other solid tumors. We performed a preliminary screen of human cancer cell lines from multiple malignancies and found that LNS8801, a synthetic pharmacologic agonist of GPER currently in early phase clinical trials, promoted apoptosis in human AML cells. Using human AML cell lines and primary cells, we show that LNS8801 inhibits human AML in preclinical in vitro models, while not affecting normal mononuclear cells. Although GPER is broadly expressed in normal and malignant myeloid cells, this cancer-specific LNS8801-induced inhibition appeared to be independent of GPER signaling. LNS8801 induced AML cell death primarily through a caspase-dependent apoptosis pathway. This was independent of secreted classical death receptor ligands, and instead required induction of reactive oxygen species (ROS) and activation of endoplasmic reticulum (ER) stress response pathways including IRE1α. These studies demonstrate a novel activity of LNS8801 in AML cells and show that targeting ER stress with LNS8801 may be a useful therapeutic approach for AML. Significance Previous work demonstrated that LNS8801 inhibits cancer via GPER activation, especially in solid tumors. Here we show that LNS8801 inhibits AML via GPER-independent mechanisms that include ROS induction and ER activation.
Collapse
Affiliation(s)
- Inyoung Lee
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Miriam Doepner
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jillian Weissenrieder
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ariana D. Majer
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sophia Mercado
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Angela Estell
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher A. Natale
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Linnaeus Therapeutics, Haddonfield, New Jersey
| | - Pamela J. Sung
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New Jersey
| | - J. Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Martin P. Carroll
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd W. Ridky
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Pardhi E, Yadav R, Chaurasiya A, Madan J, Guru SK, Singh SB, Mehra NK. Multifunctional targetable liposomal drug delivery system in the management of leukemia: Potential, opportunities, and emerging strategies. Life Sci 2023; 325:121771. [PMID: 37182551 DOI: 10.1016/j.lfs.2023.121771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/06/2023] [Accepted: 05/07/2023] [Indexed: 05/16/2023]
Abstract
The concern impeding the success of chemotherapy in leukemia treatment is descending efficacy of drugs because of multiple drug resistance (MDR). The previous failure of traditional treatment methods is primarily responsible for the present era of innovative agents to treat leukemia effectively. The treatment option is a chemotherapeutic agent in most available treatment strategies, which unfortunately leads to high unavoidable toxicities. As a result of the recent surge in marketed products, theranostic nanoparticles, i.e., multifunctional targetable liposomes (MFTL), have been approved for improved and more successful leukemia treatment that blends therapeutic and diagnostic characteristics. Since they broadly offer the required characteristics to get past the traditional/previous limitations, such as the absence of site-specific anti-cancer therapeutic delivery and ongoing real-time surveillance of the leukemia target sites while administering therapeutic activities. To prepare MFTL, suitable targeting ligands or tumor-specific antibodies are required to attach to the surface of the liposomes. This review exhaustively covered and summarized the liposomal-based formulation in leukemia treatment, emphasizing leukemia types; regulatory considerations, patents, and clinical portfolios to overcome clinical translation hurdles have all been explored.
Collapse
Affiliation(s)
- Ekta Pardhi
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Rati Yadav
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Akash Chaurasiya
- Department of Pharmaceutics, BITS-Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal, District. RR, Hyderabad, India
| | - Jitender Madan
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India.
| |
Collapse
|
31
|
Zheng X, Wu J, Song L, Huang B. ACSM3 suppresses proliferation and induces apoptosis and cell cycle arrest in acute myeloid leukemia cells via the regulation of IGF2BP2. Exp Ther Med 2023; 25:177. [PMID: 37006876 PMCID: PMC10061044 DOI: 10.3892/etm.2023.11876] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/07/2022] [Indexed: 03/11/2023] Open
Abstract
Acyl-CoA medium-chain synthetase-3 (ACSM3) has been reported to be involved in the malignant progression of multiple types of human cancer. Nevertheless, the role of ACSM3 in acute myeloid leukemia (AML) and its exact mechanism of action are as yet undefined. In the present study, the expression levels of ACSM3 and IGF2 mRNA-binding protein 2 (IGF2BP2) were evaluated using the Gene Expression Profiling Interactive Analysis database and AML cells. The Cell Counting Kit-8 assay and 5-ethynyl-2'-deoxyuridine staining were employed for the estimation of the cell proliferative activity. Induction of apoptosis and the assessment of the cell cycle were measured using flow cytometry and western blotting, respectively. The interaction of ACSM3 with IGF2BP2 was confirmed using an RNA immunoprecipitation assay. mRNA stabilization of ACSM3 following actinomycin D treatment was evaluated using reverse transcription-quantitative PCR analysis. The data indicated that the expression levels of ACSM3 were significantly downregulated, whereas those of IGF2BP2 were upregulated in tissues and AML cells. Downregulation of ACSM3 expression was closely associated with poor overall survival of patients with AML. ACSM3 overexpression repressed cell proliferative activity and induced apoptosis and cell cycle arrest. IGF2BP2 downregulated ACSM3 expression by reducing the stability of ACSM3 mRNA. In addition, IGF2BP2 overexpression counteracted the effects of ACSM3 overexpression noted on proliferation, induction of apoptosis and cell cycle arrest of HL-60 cells. In conclusion, ACSM3 repressed the cell proliferative activity and facilitated induction of apoptosis and cell cycle arrest in AML cells by modulating the expression of IGF2BP2.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Clinical Laboratory, Jianghan Oilfield General Hospital of Changjiang University, Qianjiang, Hubei 433124, P.R. China
| | - Jinjun Wu
- Department of Clinical Laboratory, Jianghan Oilfield General Hospital of Changjiang University, Qianjiang, Hubei 433124, P.R. China
| | - Linlan Song
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Bo Huang
- Department of Clinical Laboratory, The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710003, P.R. China
| |
Collapse
|
32
|
Murtadha M, Park M, Zhu Y, Caserta E, Dona AA, Singer M, Vahed H, Tasndoh T, Gonzalez A, Ly K, Sanchez JF, Chowdhury A, Pozhitkov A, Ghoda L, Li L, Zhang B, Krishnan A, Marcucci G, Williams J, Pichiorri F. Leveraging IFNγ/CD38 regulation to unmask and target leukemia stem cells in acute myelogenous leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530273. [PMID: 36909542 PMCID: PMC10002674 DOI: 10.1101/2023.02.27.530273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Elimination of drug-resistant leukemia stem cells (LSCs) represents a major challenge to achieve a cure in acute myeloid leukemia (AML). Although AML blasts generally retain high levels of surface CD38 (CD38pos), the presence of CD34 and lack of CD38 expression (CD34posCD38neg) are immunophenotypic features of both LSC-enriched AML blasts and normal hematopoietic stem cells (HSCs). We report that IFN-γ induces CD38 upregulation in LSC-enriched CD34posCD38neg AML blasts, but not in CD34posCD38neg HSCs. To leverage the IFN-γ mediated CD38 up-regulation in LSCs for clinical application, we created a compact, single-chain CD38-CD3-T cell engager (CD38-BIONIC) able to direct T cells against CD38pos blasts. Activated CD4pos and CD8pos T cells not only kill AML blasts but also produce IFNγ, which leads to CD38 expression on CD34posCD38neg LSC-enriched blasts. These cells then become CD38-BIONIC targets. The net result is an immune-mediated killing of both CD38neg and CD38pos AML blasts, which culminates in LSC depletion.
Collapse
Affiliation(s)
- Mariam Murtadha
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Miso Park
- Department of Molecular Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Yinghui Zhu
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Enrico Caserta
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Ada Alice Dona
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Mahmoud Singer
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Hawa Vahed
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Theophilus Tasndoh
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Asaul Gonzalez
- Department of Molecular Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Kevin Ly
- Department of Molecular Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - James F Sanchez
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
| | - Arnab Chowdhury
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Alex Pozhitkov
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Lucy Ghoda
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Ling Li
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Bin Zhang
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Amrita Krishnan
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
| | - Guido Marcucci
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - John Williams
- Department of Molecular Medicine, Beckman Research Institute, City of Hope; Duarte, CA, USA
| | - Flavia Pichiorri
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope; Duarte, CA, USA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope; Duarte, CA, USA
| |
Collapse
|
33
|
Calleja A, Loschi M, Bailly L, Morisot A, Marceau A, Mannone L, Robert G, Auberger P, Preudhomme C, Raynaud S, Subtil F, Sujobert P, Cluzeau T. Real-life challenges using personalized prognostic scoring systems in acute myeloid leukemia. Cancer Med 2023; 12:5656-5660. [PMID: 36394159 PMCID: PMC10028034 DOI: 10.1002/cam4.5408] [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: 07/12/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Personalized medicine is a challenge for patients with acute myeloid leukemia (AML). The identification of several genetic mutations in several AML trials led to the creation of a personalized prognostic scoring algorithm known as the Knowledge Bank (KB). In this study, we assessed the prognostic value of this algorithm on a cohort of 167 real life AML patients. We compared KB predicted outcomes to real-life outcomes. For patients younger than 60-year-old, OS was similar in favorable and intermediate ELN risk category. However, KB algorithm failed to predict OS for younger patients in the adverse ELN risk category and for patients older than 60 years old in the favorable ELN risk category. These discrepancies may be explained by the emergence of several new therapeutic options as well as the improvement of allogeneic stem cell transplantation (aHSCT) outcomes and supportive cares. Personalized medicine is a major challenge and predictions models are powerful tools to predict patient's outcome. However, the addition of new therapeutic options in the field of AML requires a prospective validation of these scoring systems to include recent therapeutic innovations.
Collapse
Affiliation(s)
- Anne Calleja
- Hematology Department, Cote D'Azur University, Nice Sophia Antipolis University, CHU of Nice, Nice, France
- Mediterranean Center for Molecular Medecine, Cote d'Azur University, INSERM U1065, Nice, France
| | - Michael Loschi
- Hematology Department, Cote D'Azur University, Nice Sophia Antipolis University, CHU of Nice, Nice, France
- Mediterranean Center for Molecular Medecine, Cote d'Azur University, INSERM U1065, Nice, France
| | - Laurent Bailly
- Public Health Department, Centre Hospitalier Universitaire de Nice, Cote d'Azur University, Nice, France
| | - Adeline Morisot
- Public Health Department, Centre Hospitalier Universitaire de Nice, Cote d'Azur University, Nice, France
| | | | - Lionel Mannone
- Hematology Department, Cote D'Azur University, Nice Sophia Antipolis University, CHU of Nice, Nice, France
| | - Guillaume Robert
- Mediterranean Center for Molecular Medecine, Cote d'Azur University, INSERM U1065, Nice, France
| | - Patrick Auberger
- Mediterranean Center for Molecular Medecine, Cote d'Azur University, INSERM U1065, Nice, France
| | - Claude Preudhomme
- Public Health Department, Centre Hospitalier Universitaire de Nice, Cote d'Azur University, Nice, France
| | - Sophie Raynaud
- Cote D'Azur University, Nice Sophia Antipolis University, CHU of Nice, Onco-Hematology Laboratory, Nice, France
| | - Fabien Subtil
- Hospices Civils de Lyon, Hôpital Lyon Sud, Service d'Hématologie Biologique, Pierre-Bénite, France
| | - Pierre Sujobert
- Hospices Civils de Lyon, Service de Biostatistique, CNRS, UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, Lyon, France
| | - Thomas Cluzeau
- Hematology Department, Cote D'Azur University, Nice Sophia Antipolis University, CHU of Nice, Nice, France
- Mediterranean Center for Molecular Medecine, Cote d'Azur University, INSERM U1065, Nice, France
| |
Collapse
|
34
|
Mendoza-Urbano DM, Tello-Cajiao ME, Rosales J, Ahumada FE, Parra-Lara LG, Arrieta E. Survival in Elderly Patients Diagnosed With Acute Myeloid Leukemia: A Hospital-Based Study. J Hematol 2023; 12:7-15. [PMID: 36895293 PMCID: PMC9990714 DOI: 10.14740/jh1055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/18/2022] [Indexed: 03/11/2023] Open
Abstract
Background Acute myeloid leukemia (AML) is a hematological neoplasm that is more frequent in elderly patients. The objective of this study was to evaluate elderly patients' survival with de novo AML and acute myeloid leukemia myelodysplasia-related (AML-MR), treated with intensive and less-intensive chemotherapy and supportive care. Methods A retrospective cohort study was conducted in Fundacion Valle del Lili (Cali, Colombia), between 2013 and 2019. We included patients ≥ 60 years old diagnosed with AML. The statistical analysis considered the leukemia type (de novo vs. myelodysplasia-related) and treatment (intensive chemotherapy regimen, less-intensive chemotherapy regimen, and without chemotherapy). Survival analysis was performed using Kaplan-Meier method and Cox regression models. Results A total of 53 patients were included (31 de novo and 22 AML-MR). Intensive chemotherapy regimens were more frequent in patients with de novo leukemia (54.8%), and 77.3% of patients with AML-MR received less-intensive regimens. Survival was higher in the chemotherapy group (P = 0.006), but with no difference between chemotherapy modalities. Additionally, patients without chemotherapy were 10 times more likely to die than those who received any regimen, independent of age, sex, Eastern Cooperative Oncology performance status, and Charlson comorbidity index (adjusted hazard ratio (HR) = 11.6, 95% confidence interval (CI) 3.47 - 38.8). Conclusions Elderly patients with AML had longer survival time when receiving chemotherapy, regardless of the type of regimen.
Collapse
Affiliation(s)
| | | | - Joaquin Rosales
- Servicio de Hemato-Oncologia, Departamento de Medicina Interna, Fundacion Valle del Lili, Cali, Colombia
| | - Fabian Emiliano Ahumada
- Servicio de Hemato-Oncologia, Departamento de Medicina Interna, Fundacion Valle del Lili, Cali, Colombia
| | - Luis Gabriel Parra-Lara
- Centro de Investigaciones Clinicas (CIC), Fundacion Valle del Lili, Cali, Colombia.,Facultad de Ciencias de la Salud, Universidad Icesi, Cali, Colombia
| | - Elizabeth Arrieta
- Servicio de Hemato-Oncologia, Departamento de Medicina Interna, Fundacion Valle del Lili, Cali, Colombia
| |
Collapse
|
35
|
Fang F, Lu J, Sang X, Tao YF, Wang JW, Zhang ZM, Zhang YP, Li XL, Xie Y, Wu SY, Chu XR, Li G, Wu D, Chen YL, Yu JJ, Jia SQ, Feng CX, Tian YY, Li ZH, Ling J, Hu SY, Pan J. Super-enhancer profiling identifies novel critical and targetable cancer survival gene LYL1 in pediatric acute myeloid leukemia. J Exp Clin Cancer Res 2022; 41:225. [PMID: 35842703 PMCID: PMC9288051 DOI: 10.1186/s13046-022-02428-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/01/2022] [Indexed: 12/26/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a myeloid neoplasm makes up 7.6% of hematopoietic malignancies. Super-enhancers (SEs) represent a special group of enhancers, which have been reported in multiple cell types. In this study, we explored super-enhancer profiling through ChIP-Seq analysis of AML samples and AML cell lines, followed by functional analysis. Methods ChIP-seq analysis for H3K27ac was performed in 11 AML samples, 7 T-ALL samples, 8 B-ALL samples, and in NB4 cell line. Genes and pathways affected by GNE-987 treatment were identified by gene expression analysis using RNA-seq. One of the genes associated with super-enhancer and affected by GNE-987 treatment was LYL1 basic helix-loop-helix family member (LYL1). shRNA mediated gene interference was used to down-regulate the expression of LYL1 in AML cell lines, and knockdown efficiency was detected by RT-qPCR and western blotting. The effect of knockdown on the growth of AML cell lines was evaluated by CCK-8. Western blotting was used to detect PARP cleavage, and flow cytometry were used to determine the effect of knockdown on apoptosis of AML cells. Results We identified a total of 200 genes which were commonly associated with super-enhancers in ≧10 AML samples, and were found enriched in regulation of transcription. Using the BRD4 inhibitor GNE-987, we assessed the dependence of AML cells on transcriptional activation for growth and found GNE-987 treatment predominantly inhibits cell growth in AML cells. Moreover, 20 candidate genes were selected by super-enhancer profile and gene expression profile and among which LYL1 was observed to promote cell growth and survival in human AML cells. Conclusions In summary, we identified 200 common super-enhancer-associated genes in AML samples, and a series of those genes are cancer genes. We also found GNE-987 treatment downregulates the expression of super-enhancer-associated genes in AML cells, including the expression of LYL1. Further functional analysis indicated that LYL1 is required for AML cell growth and survival. These findings promote understanding of AML pathophysiology and elucidated an important role of LYL1 in AML progression. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02428-9.
Collapse
|
36
|
Granroth G, Khera N, Arana Yi C. Progress and Challenges in Survivorship After Acute Myeloid Leukemia in Adults. Curr Hematol Malig Rep 2022; 17:243-253. [PMID: 36117228 PMCID: PMC9483315 DOI: 10.1007/s11899-022-00680-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Acute myeloid leukemia (AML) survivors face unique challenges affecting long-term outcomes and quality of life. There is scant literature on the long-term impact of AML treatment in physical and mental health, disease recurrence, and financial burden in survivors. RECENT FINDINGS Fatigue, mental health concerns, infections, sexual dysfunction, and increase cancer recurrence occur after AML treatment. Chronic graft-versus-host disease (GVHD) and infections are common concerns in AML after hematopoietic stem cell transplantation (HCT). Survivorship guidelines encompass symptoms and complications but fail to provide an individualized care plan for AML survivors. Studies in patient-reported outcomes (PROs) and health-related quality of life (HRQoL) are sparse. Here we discuss the most common aspects pertaining to AML survivorship, late complications, care delivery, prevention of disease recurrence, and potential areas for implementation.
Collapse
|
37
|
Anti-Leukemic Activity of Brassica-Derived Bioactive Compounds in HL-60 Myeloid Leukemia Cells. Int J Mol Sci 2022; 23:ijms232113400. [DOI: 10.3390/ijms232113400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/24/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Acute myeloid leukemia (AML) is a cancer of the myeloid blood cells mainly treated with chemotherapy for cancer remission, but this non-selective treatment also induces numerous side effects. Investigations with bioactive compounds from plant-derived foods against cancer have increased in the last years because there is an urgent need to search for new anti-leukemic agents possessing higher efficacy and selectivity for AML cells and fewer negative side effects. In this study, we analyzed the anti-leukemic activity of several phytochemicals that are representative of the major classes of compounds present in cruciferous foods (glucosinolates, isothiocyanates, hydroxycinnamic acids, flavonols, and anthocyanins) in the human acute myeloid leukemia cell line HL-60. Our results revealed that among the different Brassica-derived compounds assayed, sulforaphane (SFN) (an aliphatic isothiocyanate) showed the most potent anti-leukemic activity with an IC50 value of 6 µM in dose-response MTT assays after 48 h of treatment. On the other hand, chlorogenic acid (a hydroxycinnamic acid) and cyanidin-3-glucoside (an anthocyanin) also displayed anti-leukemic potential, with IC50 values of 7 µM and 17 µM after 48 h of incubation, respectively. Importantly, these compounds did not show significant cell toxicity in macrophages-like differentiated cells at 10 and 25 µM, indicating that their cytotoxic effects were specific to AML cancer cells. Finally, we found that these three compounds were able to induce the NRF2/KEAP1 signaling pathway in a dose-dependent manner, highlighting SFN as the most potent NRF2 activator. Overall, the present evidence shed light on the potential for using foods and ingredients rich in anticancer bioactive phytochemicals from Brassica spp.
Collapse
|
38
|
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]
|
39
|
Kubicka E, Lum LG, Huang M, Thakur A. Bispecific antibody-targeted T-cell therapy for acute myeloid leukemia. Front Immunol 2022; 13:899468. [PMID: 36389764 PMCID: PMC9663847 DOI: 10.3389/fimmu.2022.899468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/03/2022] [Indexed: 11/21/2022] Open
Abstract
The management of relapsed or refractory acute myeloid leukemia (AML) continues to be therapeutically challenging. Non-toxic immunotherapy approaches are needed to provide long-term anti-leukemic effects. The goal of this study was to determine whether activated T cells (ATCs) armed with bispecific antibodies (BiAbs) could target and lyse leukemic and leukemic stem cells (LSCs). Anti-CD3 × anti-CD123 BiAb (CD123Bi) and anti-CD3 × anti-CD33GO (gemtuzumab ozogamicin [GO]) BiAb (CD33GOBi) were used to arm ATCs to produce bispecific antibody armed activated T cells (designated CD123 BATs or CD33GO BATs) to target AML cell lines, peripheral blood mononuclear cells from AML patients, and in vivo treatment of AML in xenogeneic NSG mice engrafted with leukemic cells. BATs exhibited high levels of specific cytotoxicity directed at AML cell lines at low 1:1 or 1:2 effector-to-target (E:T) ratios and secrete Th1 cytokines upon target engagement. In vivo study in AML-engrafted NSG mice showed significantly prolonged survival in mice treated with CD33GO BATs (p < 0.0001) or CD123 BATs (p < 0.0089) compared to ATC-treated control mice. Patient samples containing leukemic blasts and LSCs when treated with CD33GO BATs or CD123 BATs for 18 h showed a significant reduction (50%-100%; p < 0.005) in blasts and 75%-100% reduction in LSCs (p < 0.005) in most cases compared to unarmed ATCs. This approach may provide a potent and non-toxic strategy to target AML blasts and LSCs and enhance chemo-responsiveness in older patients who are likely to develop recurrent diseases.
Collapse
Affiliation(s)
| | | | | | - Archana Thakur
- Cellular Immunotherapy and Bone Marrow Transplant Program, Department of Medicine, Division of Hematology/Oncology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
40
|
Xu Y, Tran L, Tang J, Nguyen V, Sewell E, Xiao J, Hino C, Wasnik S, Francis-Boyle OL, Zhang KK, Xie L, Zhong JF, Baylink DJ, Chen CS, Reeves ME, Cao H. FBP1-Altered Carbohydrate Metabolism Reduces Leukemic Viability through Activating P53 and Modulating the Mitochondrial Quality Control System In Vitro. Int J Mol Sci 2022; 23:ijms231911387. [PMID: 36232688 PMCID: PMC9570078 DOI: 10.3390/ijms231911387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML)—the most frequent form of adult blood cancer—is characterized by heterogeneous mechanisms and disease progression. Developing an effective therapeutic strategy that targets metabolic homeostasis and energy production in immature leukemic cells (blasts) is essential for overcoming relapse and improving the prognosis of AML patients with different subtypes. With respect to metabolic regulation, fructose-1,6-bisphosphatase 1 (FBP1) is a gluconeogenic enzyme that is vital to carbohydrate metabolism, since gluconeogenesis is the central pathway for the production of important metabolites and energy necessary to maintain normal cellular activities. Beyond its catalytic activity, FBP1 inhibits aerobic glycolysis—known as the “Warburg effect”—in cancer cells. Importantly, while downregulation of FBP1 is associated with carcinogenesis in major human organs, restoration of FBP1 in cancer cells promotes apoptosis and prevents disease progression in solid tumors. Recently, our large-scale sequencing analyses revealed FBP1 as a novel inducible therapeutic target among 17,757 vitamin-D-responsive genes in MV4-11 or MOLM-14 blasts in vitro, both of which were derived from AML patients with FLT3 mutations. To investigate FBP1′s anti-leukemic function in this study, we generated a new AML cell line through lentiviral overexpression of an FBP1 transgene in vitro (named FBP1-MV4-11). Results showed that FBP1-MV4-11 blasts are more prone to apoptosis than MV4-11 blasts. Mechanistically, FBP1-MV4-11 blasts have significantly increased gene and protein expression of P53, as confirmed by the P53 promoter assay in vitro. However, enhanced cell death and reduced proliferation of FBP1-MV4-11 blasts could be reversed by supplementation with post-glycolytic metabolites in vitro. Additionally, FBP1-MV4-11 blasts were found to have impaired mitochondrial homeostasis through reduced cytochrome c oxidase subunit 2 (COX2 or MT-CO2) and upregulated PTEN-induced kinase (PINK1) expressions. In summary, this is the first in vitro evidence that FBP1-altered carbohydrate metabolism and FBP1-activated P53 can initiate leukemic death by activating mitochondrial reprogramming in AML blasts, supporting the clinical potential of FBP1-based therapies for AML-like cancers.
Collapse
Affiliation(s)
- Yi Xu
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Correspondence: ; Tel.: +1-909-651-5887
| | - Lily Tran
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Janet Tang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Vinh Nguyen
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Elisabeth Sewell
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Christopher Hino
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Olivia L. Francis-Boyle
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Pathology & Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Ke K. Zhang
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA
| | - Jiang F. Zhong
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Mark E. Reeves
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| | - Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Cancer Center, Loma Linda University, Loma Linda, CA 92354, USA
| |
Collapse
|
41
|
Feasibility of Leukemia-Derived Exosome Enrichment and Co-isolated dsDNA Sequencing in Acute Myeloid Leukemia Patients: A Proof of Concept for New Leukemia Biomarkers Detection. Cancers (Basel) 2022; 14:cancers14184504. [PMID: 36139664 PMCID: PMC9497185 DOI: 10.3390/cancers14184504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary The present pilot study aimed at investigating the feasibility of a leukemia-derived exosome enrichment approach followed by exosomal dsDNA target re-sequencing for adult Acute Myeloid Leukemias (AML) marker detection. To our knowledge, this is the first time that a proof-of-concept combining a leukemia-derived exosome enrichment strategy based on a commercial CE-IVD kit and next-generation sequencing was applied in a cohort of adult AML patients. The reported approach is easy, quick and user friendly and gives the possibility of obtaining a good quantity of exosomal dsDNA (composed of exosomal cargo and surrounding DNA) suitable for further analysis. The time-effective procedure opens up future effective clinical applications. This pilot study presents the potential of a proof-of-concept based on exosome analysis to be applied in clinical practice, as well as the feasibility of this kind of investigations using a certified kit, avoiding many additional analyses. It may encourage further studies regarding extracellular vesicles in myeloid neoplasia. Abstract Exosomes are extracellular vesicles playing a pivotal role in the intercellular communication. They shuttle different cargoes, including nucleic acids from their cell of origin. For this reason, they have been studied as carriers of tumor markers in different liquid biopsy approaches, in particular for solid tumors. Few data are available concerning exosomes as markers of myeloid neoplasia. To better understand their real potential and the best approach to investigate leukemic exosomes, we present the results of a pilot feasibility study evaluating the application of next-generation sequencing analysis of dsDNA derived from exosomes isolated in 14 adult patients affected by acute myeloid leukemias. In particular, leukemia-derived exosome fractions have been analyzed. The concentration of dsDNA co-extracted with exosomes and the number and types of mutations detected were considered and compared with ones identified in the Bone Marrow (BM) and Peripheral Blood (PB) cells. Exosomal DNA concentration, both considering the cargo and the DNA surrounding the lipid membrane resulted in a linear correlation with leukemic burden. Moreover, exosomal DNA mutation status presented 86.5% of homology with BM and 75% with PB. The results of this pilot study confirmed the feasibility of a leukemia-derived exosome enrichment approach followed by exosomal dsDNA NGS analysis for AML biomarker detection. These data point to the use of liquid biopsy in myeloid neoplasia for the detection of active leukemic cells resident in the BM via a painless procedure.
Collapse
|
42
|
Chen G, She W, Yu C, Rouzi T, Li X, Ma L, Zhang N, Jiang H, Liu X, Wu J, Wang Q, Shen H, Zhou F. A novel organic arsenic derivative MZ2 remodels metabolism and triggers mtROS-mediated apoptosis in acute myeloid leukemia. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04333-2. [PMID: 36056952 DOI: 10.1007/s00432-022-04333-2] [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: 07/12/2022] [Accepted: 08/25/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Acute myeloid leukemia (AML) is one of the most common neoplasms in adults, and it is difficult to achieve satisfactory results with conventional drugs. Here, we synthesized a novel organic arsenic derivative MZ2 and evaluated its ability to remodel energy metabolism to achieve anti-leukemia. METHODS MZ2 was characterized by the average 1-min full mass spectra analysis. Biological methods such as Western blot, qPCR, flow cytometry and confocal microscopy were used to assess the mode and mechanism of MZ2-induced death. The in vivo efficacy of MZ2 was assessed by constructing a patient-derived xenograft (PDX) AML model. RESULTS Unlike the precursor organic arsenical Z2, MZ2 can effectively reduce the level of aerobic glycolysis. Our in-depth found that MZ2 inhibited the expression of PDK2 in a dose-dependent manner and did not affect the expression of LDHA, another key enzyme of the glycolytic pathway. MZ2 reconstituted energy metabolism to induce the generation of mitochondrial ROS (mtROS) and then triggerd intrinsic apoptosis pathway. We also assessed whether MZ2 generates autophagy and results showed that MZ2 can induce autophagy of AML cells, which may be associated with the precursor organic arsenic drug. In vivo, MZ2 effectively attenuated leukemia progression in mice, and immunohistochemical results suggested its PDK2 inhibitory effect. CONCLUSION In summary, the novel organic arsine derivative MZ2 exhibited excellent anti-tumor effects in acute myeloid leukemia, which may provide a potential strategy for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Guopeng Chen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Wenyan She
- College of Chemistry and Molecular Science, Wuhan University, Wuhan, 430072, Hubei, China
| | - Chaochao Yu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Tuerxunayi Rouzi
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xinqi Li
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Linlu Ma
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Nan Zhang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Hongqiang Jiang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiaoyan Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Jinxian Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Qian Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Hui Shen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
43
|
Alotaibi S, Niederwieser D, Ahmed SO, Sanz J, Mohty M, Aljurf M. Current Status of CPX-351 Therapy in Acute Myeloid Leukemia and Myelodysplastic Syndrome. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:575-580. [PMID: 35418351 DOI: 10.1016/j.clml.2022.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 06/14/2023]
Abstract
Acute myeloid leukemia (AML) treatment landscape had evolved over the last decades with better understanding of the disease genomics and the use of the targeted therapy, despite this treatment evolution, 7 + 3 remains the mainstay treatment for most AML cases. Many attempts had been made to improve the treatment outcome with 7 + 3 like manipulating the doses or the duration, but with no significant change in the outcome. In 2017 FDA approved CPX-351,a liposomal formulation of cytarabine and daunorubicin at a fixed 5:1 molar ratio, for the treatment of adults with newly diagnosed AML with myelodysplasia-related changes and therapy-related AML (t-AML). Since the approval, many trials were conducted or still ongoing in assessing the role of CPX-351 in treating different patient populations, AML subcategories or when combined with different agents. In this review, we will summarize the current role of CPX-351 in treating this largely heterogeneous disease.
Collapse
Affiliation(s)
- Shaykhah Alotaibi
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, KSA.
| | | | - Syed Osman Ahmed
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, KSA
| | - Jaime Sanz
- Hematology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Mohamad Mohty
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, Paris, France
| | - Mahmoud Aljurf
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, KSA
| |
Collapse
|
44
|
BRD4 Inhibitor GNE-987 Exerts Anticancer Effects by Targeting Super-Enhancer-Related Gene LYL1 in Acute Myeloid Leukemia. J Immunol Res 2022; 2022:7912484. [PMID: 35958877 PMCID: PMC9359861 DOI: 10.1155/2022/7912484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/17/2022] [Accepted: 07/06/2022] [Indexed: 12/13/2022] Open
Abstract
Background. AML (acute myeloid leukemia) is a common hematological malignancy in children with poor treatment effects and poor prognosis. Recent studies have shown that as a novel BRD4 (bromodomain containing 4) PROTACs (proteolysis targeting chimeras) degrader, GNE-987 can slow down the growth of various tumors and increase apoptosis, with promising clinical prospects. However, the function and molecular mechanism of GNE-987 in AML remain unclear. This study is aimed at investigating the therapeutic effect of GNE-987 on AML and its underlying mechanism. Methods. The association between BRD4 and AML was assessed by studying public databases. After GNE-987 was added to AML cells, cell proliferation slowed down, the cycle was disturbed, and apoptosis increased. Western blotting was used to detect BRD2 (bromodomain containing 2), BRD3 (bromodomain containing 3), BRD4, and PARP (poly ADP-ribose polymerase) proteins. The effect of GNE-987 on AML cells was analyzed in vivo. RNA-seq (RNA sequencing) and ChIP-seq (chromatin immunoprecipitation sequencing) validated the function and molecular pathways of GNE-987 in processing AML. Results. BRD4 expression was significantly elevated in pediatric AML samples compared with healthy donors. GNE-987 inhibited AML cell proliferation by inhibiting the cell cycle and inducing apoptosis. BRD2, BRD3, and BRD4 were consistent with decreased VHL (Von Hippel Lindau) expression in AML cells. In an AML xenograft model, GNE-987 significantly reduced the hepatosplenic infiltration of leukemia cells and increased the mouse survival time. Based on analysis of RNA-seq and ChIP-seq analyses, GNE-987 could target multiple SE- (super-enhancer-) related genes, including LYL1 (lymphoblastic leukemia 1), to inhibit AML. Conclusions. GNE-987 had strong antitumor activity in AML. GNE-987 could effectively inhibit the expression of SE-related oncogenes including LYL1 in AML. Our results suggested that GNE-987 had broad prospects in the treatment of AML.
Collapse
|
45
|
Zhao C, Wang Y, Sharma A, Wang Z, Zheng C, Wei Y, Wu Y, Liu P, Liu J, Zhan X, Schmidt-Wolf I, Tu F. Identification of the integrated prognostic signature associated with immuno-relevant genes and long non-coding RNAs in acute myeloid leukemia. Cancer Invest 2022; 40:663-674. [PMID: 35770858 DOI: 10.1080/07357907.2022.2096230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND Like other cancers, considerable effort has been made in acute myeloid leukemia (AML) to identify prognostic genes and long non-coding RNAs (lncRNAs) with their potential clinical applications. However, to date, no integrated prognostic model has been developed that combines both gene expression and lncRNAs as a singular approach in AML. METHOD Comprehensive bioinformatic approaches (Weighted gene co-expression network analysis, Univariate Cox regression analyses, Pearson correlation, LASSO-Cox regression, Wilcoxon test) were used to construct the signature and to define high- and low-risk groups in AML datasets. ESTIMATE and CIBERSORT algorithms were applied to investigate the potential impact of infiltrating immune cells based on the obtained signature in tumor microenvironment. In addition, gene ontology (GO) and KEGG enrichment were applied to explore the potential function of the signature. RESULTS Herein, we focused on immune-related genes (IRGs) and immune-related long non-coding RNAs (IRlncRNAs) and constructed an integrated prognostic immunorelevant signature in AML. The obtained signature exhibit five IRGs (DAXX, PSMB8, CSRP1, RAC2 and PTPN6) and one IRlncRNA (AC080037.2), and is strictly associated with age and FAB (French-American-British classification). Importantly, the high-risk AML group (defined by the signature) correlated positively with three types of scores (immune score, stroma score, and ESTIMATE score). We also identified a few immune cells (resting mast cells and monocytes) potentially involved in the correlation between signature and survival of AML patients. The prognostic ability of the obtained signature was tested in the training cohort and then validated in both test and total cohorts. The pathway enrichment analysis confirmed the possible immune- related role of the signature. CONCLUSION We constructed an integrated prognostic signature comprising five immune-related protein-coding genes (IRPCG) (DAXX, PSMB8, CSRP1, RAC2, and PTPN6) and one immune-related lncRNA (AC080037.2) that may serve as potential biomarkers for predicting survival and further stratifying AML patients.
Collapse
Affiliation(s)
- Chunxia Zhao
- Department of Nursing, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China.,School of Nursing, Nanchang University, Nanchang 330006, China
| | - Yulu Wang
- Department of Integrated Oncology, Center for Integrated Oncology, University Hospital Bonn, Bonn 53127, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology, University Hospital Bonn, Bonn 53127, Germany.,Department of Neurosurgery, University Hospital Bonn, Bonn 53127, Germany
| | - Zifeng Wang
- Department of Hematology, Shangrao people's hospital, Shangrao 334000, China
| | - Chafeng Zheng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ying Wei
- Department of Pathology, Shangrao people's hospital, Shangrao 334000, China
| | - Yun Wu
- Department of Hematology, Shangrao people's hospital, Shangrao 334000, China
| | - Pingping Liu
- Department of Nursing, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jiachen Liu
- School of Nursing, Nanchang University, Nanchang 330006, China
| | - Xulong Zhan
- Department of Hematology,The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Ingo Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology, University Hospital Bonn, Bonn 53127, Germany
| | - Famei Tu
- Department of Nursing, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
46
|
Zeng P, Chai Y, You C, Yue L, Wu C, Chen H, Li L, Li J, Liu H, Zhang Y, Cao T, Li Y, Hu W. Long Noncoding RNA Small Nucleolar RNA Host gene 1 is not Only Overexpressed, But Also Correlates with Increased White Blood Cell Count, Deteriorate Induction Treatment Response and Poor Survival Profile in Adult Acute Myeloid Leukemia Patients. Turk J Haematol 2022; 39:188-195. [PMID: 35657175 PMCID: PMC9421341 DOI: 10.4274/tjh.galenos.2022.2022.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective Long non-coding RNA small nucleolar RNA host gene 1 (lnc-SNHG1) is involved in leukemogenesis via mediating multiple pathways. The current study aimed to further explore its clinical role concerning disease risk, clinical features, and prognostication in patients with acute myeloid leukemia (AML). Material and Methods Generally, 161 adult AML patients, 50 disease controls (DC), and 50 healthy controls (HC) were enrolled; bone marrow mononuclear cells (BMMCs) were collected. Subsequently, a reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to measure the lnc-SNHG1 expression. Results Lnc-SNHG1 expression was higher in AML patients than that in DC and HC (both P<0.001), with a good value in distinguishing AML patients from DC and HC (area under the curve (AUC) of 0.726 and 0.884, respectively). Moreover, lnc-SNHG1 expression was positively associated with white blood cell (WBC) count (P=0.008) but not correlated with other clinical features such as cytogenetics, molecule-genetics, and risk stratifications, etc. (all P>0.05). Lnc-SNHG1 expression was associated with lower complete remission (CR) rate (P=0.001). In detail, patients with lnc-SNHG1 expression at quantile-4 had the worst CR rate compared to patients with lnc-SNHG1 expression at quantile-1, quantile-2, and quantile-3 (all P<0.05). Besides, lnc-SNHG1 expression was correlated with unsatisfied event-free survival (P<0.001) and overall survival (P=0.002), which were worst in patients with lnc-SNHG1 expression at quantile-4 compared with patients with lnc-SNHG1 expression at quantile-1, quantile-2 and quantile-3 (all P<0.05). Conclusion Lnc-SNHG1 overexpression is associated with elevated WBC, deteriorated induction treatment response, and poor survival profile in AML patients, which would further serve as a potential indicator for AML.
Collapse
Affiliation(s)
- Pengyun Zeng
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Ye Chai
- Department of Clinical Laboratory, Lanzhou University, Second Hospital, Lanzhou, China
| | - Chongge You
- Department of Clinical Laboratory, Lanzhou University, Second Hospital, Lanzhou, China
| | - Lingling Yue
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Chongyang Wu
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Huiling Chen
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Liangliang Li
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Jingjing Li
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Huan Liu
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Yurong Zhang
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Tingyong Cao
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Yaru Li
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| | - Wanli Hu
- Department of Hematology, Lanzhou University, Second Hospital, Lanzhou, China
| |
Collapse
|
47
|
Liu Z, Zhong L, Dan W, Chu X, Liu C, Luo X, Zhang Z, Lu Y, Wan P, Wang X, Liu B. miRNA-222-3p enhances the proliferation and suppresses the apoptosis of acute myeloid leukemia cells by targeting Axin2 and modulating the Wnt/β-catenin pathway. Biochem Biophys Res Commun 2022; 620:83-91. [DOI: 10.1016/j.bbrc.2022.06.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/17/2022] [Indexed: 11/29/2022]
|
48
|
Cao H, Tadros V, Hiramoto B, Leeper K, Hino C, Xiao J, Pham B, Kim DH, Reeves ME, Chen CS, Zhong JF, Zhang KK, Xie L, Wasnik S, Baylink DJ, Xu Y. Targeting TKI-Activated NFKB2-MIF/CXCLs-CXCR2 Signaling Pathways in FLT3 Mutated Acute Myeloid Leukemia Reduced Blast Viability. Biomedicines 2022; 10:biomedicines10051038. [PMID: 35625776 PMCID: PMC9138861 DOI: 10.3390/biomedicines10051038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
Disease relapse is a common cause of treatment failure in FMS-like tyrosine kinase 3 (FLT3) mutated acute myeloid leukemia (AML). In this study, to identify therapeutic targets responsible for the survival and proliferation of leukemic cells (blasts) with FLT3 mutations after gilteritinib (GILT, a 2nd generation tyrosine kinase inhibitor (TKI)) treatment, we performed proteomic screening of cytokine release and in vitro/ex vivo studies to investigate their associated signaling pathways and transcriptional regulation. Here, we report that macrophage migration inhibition factor (MIF) was significantly increased in the supernatant of GILT-treated blasts when compared to untreated controls. Additionally, the GILT-treated blasts that survived were found to exhibit higher expressions of the CXCR2 gene and protein, a common receptor for MIF and pro-inflammatory cytokines. The supplementation of exogenous MIF to GILT-treated blasts revealed a group of CD44High+ cells that might be responsible for the relapse. Furthermore, we identified the highly activated non-classical NFKB2 pathway after GILT-treatment. The siRNA transient knockdown of NFKB2 significantly reduced the gene expressions of MIF, CXCR2, and CXCL5. Finally, treatments of AML patient samples ex vivo demonstrated that the combination of a pharmaceutical inhibitor of the NFKB family and GILT can effectively suppress primary blasts’ secretion of tumor-promoting cytokines, such as CXCL1/5/8. In summary, we provide the first evidence that targeting treatment-activated compensatory pathways, such as the NFKB2-MIF/CXCLs-CXCR2 axis could be a novel therapeutic strategy to overcome TKI-resistance and effectively treat AML patients with FLT3 mutations.
Collapse
Affiliation(s)
- Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (H.C.); (C.H.); (B.P.); (M.E.R.); (C.-S.C.)
- Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
| | - Verena Tadros
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (V.T.); (B.H.); (K.L.); (J.X.); (D.H.K.); (S.W.); (D.J.B.)
| | - Benjamin Hiramoto
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (V.T.); (B.H.); (K.L.); (J.X.); (D.H.K.); (S.W.); (D.J.B.)
| | - Kevin Leeper
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (V.T.); (B.H.); (K.L.); (J.X.); (D.H.K.); (S.W.); (D.J.B.)
| | - Christopher Hino
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (H.C.); (C.H.); (B.P.); (M.E.R.); (C.-S.C.)
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (V.T.); (B.H.); (K.L.); (J.X.); (D.H.K.); (S.W.); (D.J.B.)
| | - Bryan Pham
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (H.C.); (C.H.); (B.P.); (M.E.R.); (C.-S.C.)
| | - Do Hyun Kim
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (V.T.); (B.H.); (K.L.); (J.X.); (D.H.K.); (S.W.); (D.J.B.)
| | - Mark E. Reeves
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (H.C.); (C.H.); (B.P.); (M.E.R.); (C.-S.C.)
- Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (H.C.); (C.H.); (B.P.); (M.E.R.); (C.-S.C.)
- Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
| | - Jiang F. Zhong
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA;
| | - Ke K. Zhang
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA; (K.K.Z.); (L.X.)
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77030, USA; (K.K.Z.); (L.X.)
| | - Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (V.T.); (B.H.); (K.L.); (J.X.); (D.H.K.); (S.W.); (D.J.B.)
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (V.T.); (B.H.); (K.L.); (J.X.); (D.H.K.); (S.W.); (D.J.B.)
| | - Yi Xu
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (H.C.); (C.H.); (B.P.); (M.E.R.); (C.-S.C.)
- Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (V.T.); (B.H.); (K.L.); (J.X.); (D.H.K.); (S.W.); (D.J.B.)
- Correspondence: ; Tel.: +1-9096515887
| |
Collapse
|
49
|
Xu Y, Hino C, Baylink DJ, Xiao J, Reeves ME, Zhong JF, Mirshahidi S, Cao H. Vitamin D activates FBP1 to block the Warburg effect and modulate blast metabolism in acute myeloid leukemia. Biomark Res 2022; 10:16. [PMID: 35366947 PMCID: PMC8977002 DOI: 10.1186/s40364-022-00367-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/18/2022] [Indexed: 12/21/2022] Open
Abstract
AbstractAcute myeloid leukemia (AML) has the lowest survival rate among the leukemias. Targeting intracellular metabolism and energy production in leukemic cells can be a promising therapeutic strategy for AML. Recently, we presented the successful use of vitamin D (1,25VD3) gene therapy to treat AML mouse models in vivo. In this study, recognizing the importance of 1,25VD3 as one of only 2 molecules (along with glucose) photosynthesized for energy during the beginning stage of life on this planet, we explored the functional role of 1,25VD3 in AML metabolism.Transcriptome database (RNA-seq) of four different AML cell lines revealed 17,757 genes responding to 1,25VD3-treatment. Moreover, we discovered that fructose-bisphosphatase 1 (FBP1) noticeably stands out as the only gene (out of 17,757 genes) with a 250-fold increase in gene expression, which is known to encode the key rate-limiting gluconeogenic enzyme fructose-1,6-bisphosphatase. The significant increased expression of FBP1 gene and proteins induced by 1,25VD3 was confirmed by qPCR, western blot, flow cytometry, immunocytochemistry and functional lactate assay. Additionally, 1,25VD3 was found to regulate different AML metabolic processes including gluconeogenesis, glycolysis, TCA, de novo nucleotide synthesis, etc. In summary, we provided the first evidence that 1,25 VD3-induced FBP1 overexpression might be a novel therapeutic target to block the “Warburg Effect” to reduce energy production in AML blasts.
Collapse
|
50
|
Artesunate improves venetoclax plus cytarabine AML cell targeting by regulating the Noxa/Bim/Mcl-1/p-Chk1 axis. Cell Death Dis 2022; 13:379. [PMID: 35443722 PMCID: PMC9021233 DOI: 10.1038/s41419-022-04810-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 11/17/2022]
Abstract
Venetoclax plus cytarabine therapy is approved for elderly acute myeloid leukemia (AML) patients and needs further improvement. We studied the mechanisms of venetoclax plus cytarabine treatment and searched for a third agent to enhance their effects. Cytarabine induces S phase arrest-mediated DNA damage with activation of DNA replication checkpoint kinase 1 (Chk1) through phosphorylation, while venetoclax induces B cell lymphoma 2 (Bcl-2)-interacting mediator of cell death (Bim)-mediated apoptotic DNA damage. Myeloid cell leukemia-1 (Mcl-1) plays negative roles in both events by sequestering Bim and accelerating Chk1 phosphorylation. Venetoclax releases Bim from Bcl-2 with increased Bim binding to Mcl-1. Artesunate, an antimalaria drug, induces Noxa to replace Bim from Mcl-1 and induces synergistic apoptosis with venetoclax accompanied with Mcl-1 reduction. Silencing Mcl-1 or adding venetoclax/artesunate diminishes the cytarabine resistance pathway p-Chk1. The triple combination exhibits S phase arrest with enhanced DNA damage, improves AML colony formation inhibition, and prolongs survival of two mice xenograft models compared to the venetoclax/cytarabine dual combination. Artesunate serves as a bridge for venetoclax and cytarabine combination by Noxa and Bim-mediated apoptosis and Mcl-1 reduction. We provide a new triple combination for AML treatment by targeting the Noxa/Mcl-1/Bim axis to reverse Mcl-1/p-Chk1 resistance of cytarabine therapy.
Collapse
|