101
|
Zhang T, Liu C, Li W, Kuang J, Qiu XY, Min L, Zhu L. Targeted protein degradation in mammalian cells: A Promising Avenue toward Future. Comput Struct Biotechnol J 2022; 20:5477-5489. [PMID: 36249565 PMCID: PMC9535385 DOI: 10.1016/j.csbj.2022.09.038] [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: 06/16/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 12/04/2022] Open
Abstract
In the eukaryotic cellular milieu, proteins are continuously synthesized and degraded effectively via endogenous protein degradation machineries such as the ubiquitin–proteasome and lysosome pathways. By reengineering and repurposing these natural protein regulatory mechanisms, the targeted protein degradation (TPD) strategies are presenting biologists with powerful tools to manipulate the abundance of proteins of interest directly, precisely, and reversibly at the post-translational level. In recent years, TPD is gaining massive attention and is recognized as a paradigm shift both in basic research, application-oriented synthetic biology, and pioneering clinical work. In this review, we summarize the updated information, especially the engineering efforts and developmental route, of current state-of-the-art TPD technology such as Trim-Away, LYTACs, and AUTACs. Besides, the general design principle, benefits, problems, and opportunities to be addressed were further analyzed, with the aim of providing guidelines for exploration, discovery, and further application of novel TPD tools in the future.
Collapse
|
102
|
Prolonged XPO1 inhibition is essential for optimal antileukemic activity in NPM1-mutated AML. Blood Adv 2022; 6:5938-5949. [PMID: 36037515 PMCID: PMC9701620 DOI: 10.1182/bloodadvances.2022007563] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/10/2022] [Indexed: 12/15/2022] Open
Abstract
NPM1 is the most frequently mutated gene in adults with acute myeloid leukemia (AML). The interaction between mutant NPM1 (NPM1c) and exportin-1 (XPO1) causes aberrant cytoplasmic dislocation of NPM1c and promotes the high expression of homeobox (HOX) genes, which is critical for maintaining the leukemic state of NPM1-mutated cells. Although there is a rationale for using XPO1 inhibitors in NPM1-mutated AML, selinexor administered once or twice per week did not translate into clinical benefit in patients with NPM1 mutations. Here, we show that this dosing strategy results in only a temporary disruption of the XPO1-NPM1c interaction, limiting the efficacy of selinexor. Because the second-generation XPO1 inhibitor eltanexor can be administered more frequently, we tested the antileukemic activity of prolonged XPO1 inhibition in NPM1-mutated AML models. Eltanexor caused irreversible HOX downregulation, induced terminal AML differentiation, and prolonged the survival of leukemic mice. This study provides essential information for the appropriate design of clinical trials with XPO1 inhibitors in NPM1-mutated AML.
Collapse
|
103
|
Zhang HB, Sun ZK, Zhong FM, Yao FY, Liu J, Zhang J, Zhang N, Lin J, Li SQ, Li MY, Jiang JY, Cheng Y, Xu S, Cheng XX, Huang B, Wang XZ. A novel fatty acid metabolism-related signature identifies features of the tumor microenvironment and predicts clinical outcome in acute myeloid leukemia. Lipids Health Dis 2022; 21:79. [PMID: 36002858 PMCID: PMC9404605 DOI: 10.1186/s12944-022-01687-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is the most common malignancy of the hematological system, and there are currently a number of studies regarding abnormal alterations in energy metabolism, but fewer reports related to fatty acid metabolism (FAM) in AML. We therefore analyze the association of FAM and AML tumor development to explore targets for clinical prognosis prediction and identify those with potential therapeutic value. Methods The identification of AML patients with different fatty acid metabolism characteristics was based on a consensus clustering algorithm. The CIBERSORT algorithm was used to calculate the proportion of infiltrating immune cells. We used Cox regression analysis and least absolute shrinkage and selection operator (LASSO) regression analysis to construct a signature for predicting the prognosis of AML patients. The Genomics of Drug Sensitivity in Cancer database was used to predict the sensitivity of patient samples in high- and low-risk score groups to different chemotherapy drugs. Results The consensus clustering approach identified three molecular subtypes of FAM that exhibited significant differences in genomic features such as immunity, metabolism, and inflammation, as well as patient prognosis. The risk-score model we constructed accurately predicted patient outcomes, with area under the receiver operating characteristic curve values of 0.870, 0.878, and 0.950 at 1, 3, and 5 years, respectively. The validation cohort also confirmed the prognostic evaluation performance of the risk score. In addition, higher risk scores were associated with stronger fatty acid metabolisms, significantly higher expression levels of immune checkpoints, and significantly increased infiltration of immunosuppressive cells. Immune functions, such as inflammation promotion, para-inflammation, and type I/II interferon responses, were also significantly activated. These results demonstrated that immunotherapy targeting immune checkpoints and immunosuppressive cells, such as myeloid-derived suppressor cells (MDSCs) and M2 macrophages, are more suitable for patients with high-risk scores. Finally, the prediction results of chemotherapeutic drugs showed that samples in the high-risk score group had greater treatment sensitivity to four chemotherapy drugs in vitro. Conclusions The analysis of the molecular patterns of FAM effectively predicted patient prognosis and revealed various tumor microenvironment (TME) characteristics. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-022-01687-x.
Collapse
Affiliation(s)
- Hai-Bin Zhang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhuo-Kai Sun
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Fang-Min Zhong
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Fang-Yi Yao
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jing Liu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jing Zhang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Nan Zhang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jin Lin
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shu-Qi Li
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Mei-Yong Li
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jun-Yao Jiang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ying Cheng
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Shuai Xu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Xue-Xin Cheng
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Xiao-Zhong Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China. .,School of Public Health, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
104
|
Current status and future perspectives in targeted therapy of NPM1-mutated AML. Leukemia 2022; 36:2351-2367. [PMID: 36008542 PMCID: PMC9522592 DOI: 10.1038/s41375-022-01666-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/09/2022]
Abstract
Nucleophosmin 1 (NPM1) is a nucleus-cytoplasmic shuttling protein which is predominantly located in the nucleolus and exerts multiple functions, including regulation of centrosome duplication, ribosome biogenesis and export, histone assembly, maintenance of genomic stability and response to nucleolar stress. NPM1 mutations are the most common genetic alteration in acute myeloid leukemia (AML), detected in about 30–35% of adult AML and more than 50% of AML with normal karyotype. Because of its peculiar molecular and clinico-pathological features, including aberrant cytoplasmic dislocation of the NPM1 mutant and wild-type proteins, lack of involvement in driving clonal hematopoiesis, mutual exclusion with recurrent cytogenetic abnormalities, association with unique gene expression and micro-RNA profiles and high stability at relapse, NPM1-mutated AML is regarded as a distinct genetic entity in the World Health Organization (WHO) classification of hematopoietic malignancies. Starting from the structure and functions of NPM1, we provide an overview of the potential targeted therapies against NPM1-mutated AML and discuss strategies aimed at interfering with the oligomerization (compound NSC348884) and the abnormal traffic of NPM1 (avrainvillamide, XPO1 inhibitors) as well as at inducing selective NPM1-mutant protein degradation (ATRA/ATO, deguelin, (-)-epigallocatechin-3-gallate, imidazoquinoxaline derivatives) and at targeting the integrity of nucleolar structure (actinomycin D). We also discuss the current therapeutic results obtained in NPM1-mutated AML with the BCL-2 inhibitor venetoclax and the preliminary clinical results using menin inhibitors targeting HOX/MEIS1 expression. Finally, we review various immunotherapeutic approaches in NPM1-mutated AML, including immune check-point inhibitors, CAR and TCR T-cell-based therapies against neoantigens created by the NPM1 mutations.
Collapse
|
105
|
Meyer AE, Stelloh C, Pulakanti K, Burns R, Fisher JB, Heimbruch KE, Tarima S, Furumo Q, Brennan J, Zheng Y, Viny AD, Vassiliou GS, Rao S. Combinatorial genetics reveals the Dock1-Rac2 axis as a potential target for the treatment of NPM1;Cohesin mutated AML. Leukemia 2022; 36:2032-2041. [PMID: 35778533 PMCID: PMC9357218 DOI: 10.1038/s41375-022-01632-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 02/03/2023]
Abstract
Acute myeloid leukemia (AML) is driven by mutations that occur in numerous combinations. A better understanding of how mutations interact with one another to cause disease is critical to developing targeted therapies. Approximately 50% of patients that harbor a common mutation in NPM1 (NPM1cA) also have a mutation in the cohesin complex. As cohesin and Npm1 are known to regulate gene expression, we sought to determine how cohesin mutation alters the transcriptome in the context of NPM1cA. We utilized inducible Npm1cAflox/+ and core cohesin subunit Smc3flox/+ mice to examine AML development. While Npm1cA/+;Smc3Δ/+ mice developed AML with a similar latency and penetrance as Npm1cA/+ mice, RNA-seq suggests that the Npm1cA/+; Smc3Δ/+ mutational combination uniquely alters the transcriptome. We found that the Rac1/2 nucleotide exchange factor Dock1 was specifically upregulated in Npm1cA/+;Smc3Δ/+ HSPCs. Knockdown of Dock1 resulted in decreased growth and adhesion and increased apoptosis only in Npm1cA/+;Smc3Δ/+ AML. Higher Rac activity was also observed in Npm1cA/+;Smc3Δ/+ vs. Npm1cA/+ AMLs. Importantly, the Dock1/Rac pathway is targetable in Npm1cA/+;Smc3Δ/+ AMLs. Our results suggest that Dock1/Rac represents a potential target for the treatment of patients harboring NPM1cA and cohesin mutations and supports the use of combinatorial genetics to identify novel precision oncology targets.
Collapse
Affiliation(s)
| | - Cary Stelloh
- Blood Research Institute, Versiti, Milwaukee, WI, USA
| | | | - Robert Burns
- Blood Research Institute, Versiti, Milwaukee, WI, USA
| | - Joseph B Fisher
- Department of Natural Sciences, Concordia University Wisconsin, Mequon, WI, USA
| | - Katelyn E Heimbruch
- Blood Research Institute, Versiti, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sergey Tarima
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Quinlan Furumo
- Department of Biology, Boston College, Chestnut Hill, MA, USA
| | - John Brennan
- Department of Pathology and Lab Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Yongwei Zheng
- Guangzhou Bio-gene Technology Co., Ltd., Guangzhou, China
| | - Aaron D Viny
- Department of Medicine, Division of Hematology and Oncology and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - George S Vassiliou
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
| | - Sridhar Rao
- Blood Research Institute, Versiti, Milwaukee, WI, USA.
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Pediatrics, Division of Hematology, Oncology, and Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
106
|
Griffioen MS, de Leeuw DC, Janssen JJWM, Smit L. Targeting Acute Myeloid Leukemia with Venetoclax; Biomarkers for Sensitivity and Rationale for Venetoclax-Based Combination Therapies. Cancers (Basel) 2022; 14:cancers14143456. [PMID: 35884517 PMCID: PMC9318140 DOI: 10.3390/cancers14143456] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Venetoclax has proven to be a promising therapy for newly diagnosed, relapsed and refractory AML patients ineligible for induction chemotherapy. Current ongoing clinical trials are evaluating its effectivity as frontline therapy for all acute myeloid leukemia (AML) patients. However, response rates vary wildly, depending on patient characteristics and mutational profiles. This review elaborates on the efficacy and safety of venetoclax compared to conventional chemotherapy for treatment of AML patients, comparing the response rates, overall survival and adverse events. Moreover, it gives an overview of genetic and epigenetic AML cell characteristics that give enhanced or decreased response to venetoclax and offers insights into the pathogenesis of venetoclax sensitivity and resistance. Additionally, it suggests possible treatment combinations predicted to be successful based on identified mechanisms influencing venetoclax sensitivity of AML cells. Abstract Venetoclax is a BCL-2 inhibitor that effectively improves clinical outcomes in newly diagnosed, relapsed and refractory acute myeloid leukemia (AML) patients, with complete response rates (with and without complete blood count recovery) ranging between 34–90% and 21–33%, respectively. Here, we aim to give an overview of the efficacy of venetoclax-based therapy for AML patients, as compared to standard chemotherapy, and on factors and mechanisms involved in venetoclax sensitivity and resistance in AML (stem) cells, with the aim to obtain a perspective of response biomarkers and combination therapies that could enhance the sensitivity of AML cells to venetoclax. The presence of molecular aberrancies can predict responses to venetoclax, with a higher response in NPM1-, IDH1/2-, TET2- and relapsed or refractory RUNX1-mutated AML. Decreased sensitivity to venetoclax was observed in patients harboring FLT3-ITD, TP53, K/NRAS or PTPN11 mutations. Moreover, resistance to venetoclax was observed in AML with a monocytic phenotype and patients pre-treated with hypomethylating agents. Resistance to venetoclax can arise due to mutations in BCL-2 or pro-apoptotic proteins, an increased dependency on MCL-1, and usage of additional/alternative sources for energy metabolism, such as glycolysis and fatty acid metabolism. Clinical studies are testing combination therapies that may circumvent resistance, including venetoclax combined with FLT3- and MCL-1 inhibitors, to enhance venetoclax-induced cell death. Other treatments that can potentially synergize with venetoclax, including MEK1/2 and mitochondrial complex inhibitors, need to be evaluated in a clinical setting.
Collapse
Affiliation(s)
- Mila S Griffioen
- Department of Hematology, Amsterdam UMC, Location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - David C de Leeuw
- Department of Hematology, Amsterdam UMC, Location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Jeroen J W M Janssen
- Department of Hematology, Amsterdam UMC, Location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Linda Smit
- Department of Hematology, Amsterdam UMC, Location VUmc, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
107
|
Khan I, Gartel AL. The antagonistic duality of NPM1 mutations in AML. Blood Adv 2022; 6:4028-4030. [PMID: 35507748 PMCID: PMC9278292 DOI: 10.1182/bloodadvances.2022007420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/01/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Irum Khan
- Division of Hematology and Oncology, Department of Medicine, and
| | - Andrei L. Gartel
- Division of Hepatology, Department of Medicine, University of Illinois, Chicago, IL
| |
Collapse
|
108
|
Lin KH, Rutter JC, Xie A, Killarney ST, Vaganay C, Benaksas C, Ling F, Sodaro G, Meslin PA, Bassil CF, Fenouille N, Hoj J, Washart R, Ang HX, Cerda-Smith C, Chaintreuil P, Jacquel A, Auberger P, Forget A, Itzykson R, Lu M, Lin J, Pierobon M, Sheng Z, Li X, Chilkoti A, Owzar K, Rizzieri DA, Pardee TS, Benajiba L, Petricoin E, Puissant A, Wood KC. P2RY2-AKT activation is a therapeutically actionable consequence of XPO1 inhibition in acute myeloid leukemia. NATURE CANCER 2022; 3:837-851. [PMID: 35668193 PMCID: PMC9949365 DOI: 10.1038/s43018-022-00394-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022]
Abstract
Selinexor is a first-in-class inhibitor of the nuclear exportin XPO1 that was recently approved by the US Food and Drug Administration for the treatment of multiple myeloma and diffuse large B-cell lymphoma. In relapsed/refractory acute myeloid leukemia (AML), selinexor has shown promising activity, suggesting that selinexor-based combination therapies may have clinical potential. Here, motivated by the hypothesis that selinexor's nuclear sequestration of diverse substrates imposes pleiotropic fitness effects on AML cells, we systematically catalog the pro- and anti-fitness consequences of selinexor treatment. We discover that selinexor activates PI3Kγ-dependent AKT signaling in AML by upregulating the purinergic receptor P2RY2. Inhibiting this axis potentiates the anti-leukemic effects of selinexor in AML cell lines, patient-derived primary cultures and multiple mouse models of AML. In a syngeneic, MLL-AF9-driven mouse model of AML, treatment with selinexor and ipatasertib outperforms both standard-of-care chemotherapy and chemotherapy with selinexor. Together, these findings establish drug-induced P2RY2-AKT signaling as an actionable consequence of XPO1 inhibition in AML.
Collapse
Affiliation(s)
- Kevin H Lin
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Justine C Rutter
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Abigail Xie
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Shane T Killarney
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Camille Vaganay
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Chaima Benaksas
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Frank Ling
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Gaetano Sodaro
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Paul-Arthur Meslin
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | | | - Nina Fenouille
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Jacob Hoj
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Rachel Washart
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Hazel X Ang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | | | | | | | | | - Antoine Forget
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Raphael Itzykson
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Min Lu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Jiaxing Lin
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Mariaelena Pierobon
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Zhecheng Sheng
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Xinghai Li
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - David A Rizzieri
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Timothy S Pardee
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Lina Benajiba
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France
| | - Emanuel Petricoin
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Alexandre Puissant
- Université de Paris, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Paris, France.
| | - Kris C Wood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| |
Collapse
|
109
|
Pardieu B, Pasanisi J, Ling F, Dal Bello R, Penneroux J, Su A, Joudinaud R, Chat L, Wu HC, Duchmann M, Sodaro G, Chauvel C, Castelli FA, Vasseur L, Pacchiardi K, Belloucif Y, Laiguillon MC, Meduri E, Vaganay C, Alexe G, Berrou J, Benaksas C, Forget A, Braun T, Gardin C, Raffoux E, Clappier E, Adès L, de Thé H, Fenaille F, Huntly BJ, Stegmaier K, Dombret H, Fenouille N, Lobry C, Puissant A, Itzykson R. Cystine uptake inhibition potentiates front-line therapies in acute myeloid leukemia. Leukemia 2022; 36:1585-1595. [PMID: 35474100 DOI: 10.1038/s41375-022-01573-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 12/17/2022]
Abstract
By querying metabolic pathways associated with leukemic stemness and survival in multiple AML datasets, we nominated SLC7A11 encoding the xCT cystine importer as a putative AML dependency. Genetic and chemical inhibition of SLC7A11 impaired the viability and clonogenic capacity of AML cell lines in a cysteine-dependent manner. Sulfasalazine, a broadly available drug with xCT inhibitory activity, had anti-leukemic activity against primary AML samples in ex vivo cultures. Multiple metabolic pathways were impacted upon xCT inhibition, resulting in depletion of glutathione pools in leukemic cells and oxidative stress-dependent cell death, only in part through ferroptosis. Higher expression of cysteine metabolism genes and greater cystine dependency was noted in NPM1-mutated AMLs. Among eight anti-leukemic drugs, the anthracycline daunorubicin was identified as the top synergistic agent in combination with sulfasalazine in vitro. Addition of sulfasalazine at a clinically relevant concentration significantly augmented the anti-leukemic activity of a daunorubicin-cytarabine combination in a panel of 45 primary samples enriched in NPM1-mutated AML. These results were confirmed in vivo in a patient-derived xenograft model. Collectively, our results nominate cystine import as a druggable target in AML and raise the possibility to repurpose sulfasalazine for the treatment of AML, notably in combination with chemotherapy.
Collapse
Affiliation(s)
- Bryann Pardieu
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Justine Pasanisi
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Frank Ling
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Reinaldo Dal Bello
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Justine Penneroux
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Angela Su
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Romane Joudinaud
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Laureen Chat
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Hsin Chieh Wu
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR1050, CNRS UMR, 7241, Paris, France
| | - Matthieu Duchmann
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Gaetano Sodaro
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Clémentine Chauvel
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Florence A Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif-sur-Yvette, France
| | - Loic Vasseur
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Kim Pacchiardi
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Yannis Belloucif
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Marie-Charlotte Laiguillon
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Eshwar Meduri
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - Camille Vaganay
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Gabriela Alexe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeannig Berrou
- Université Paris Cité, Leukemia Transfer Lab, EA 3518, Institut de Recherche Saint-Louis, F-75010, Paris, France
| | - Chaima Benaksas
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Antoine Forget
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Thorsten Braun
- Université Paris Cité, Leukemia Transfer Lab, EA 3518, Institut de Recherche Saint-Louis, F-75010, Paris, France
| | - Claude Gardin
- Université Paris Cité, Leukemia Transfer Lab, EA 3518, Institut de Recherche Saint-Louis, F-75010, Paris, France
| | - Emmanuel Raffoux
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Emmanuelle Clappier
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Lionel Adès
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
| | - Hugues de Thé
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR1050, CNRS UMR, 7241, Paris, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, F-91191, Gif-sur-Yvette, France
| | - Brian J Huntly
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hervé Dombret
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France
- The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nina Fenouille
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Camille Lobry
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Alexandre Puissant
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France
| | - Raphael Itzykson
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, F-75010, Paris, France.
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, F-75010, Paris, France.
| |
Collapse
|
110
|
Shi Y, Xue Y, Wang C, Yu L. Nucleophosmin 1: from its pathogenic role to a tantalizing therapeutic target in acute myeloid leukemia. Hematology 2022; 27:609-619. [PMID: 35621728 DOI: 10.1080/16078454.2022.2067939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Nucleophosmin 1 (NPM1, also known as B23) is a multifunctional protein involved in a variety of cellular processes, including ribosomal maturation, centrosome replication, maintenance of genomic stability, cell cycle control, and apoptosis. NPM1 is the most commonly mutated gene in adult acute myeloid leukemia (AML) and is present in approximately 40% of all AML cases. The underlying mechanisms of mutant NPM1 (NPM1mut) in leukemogenesis remain unclear. This review summarizes the structure and physiological function of NPM1, mechanisms underlying the pathogenesis of NPM1-mutated AML, and the potential role of NPM1 as a therapeutic target. It is reported that dysfunctional NPM1 might cause AML pathogenesis via its role as a protein chaperone, inhibiting differentiation of leukemia stem cells and regulation of non-coding RNAs. Besides conventional chemotherapies, NPM1 is a promising therapeutic target against AML that warrants further investigation. NPM1-based therapeutic strategies include inducing nucleolar relocalisation of NPM1 mutants, interfering with NPM1 oligomerization, and NPM1 as an immune response target.
Collapse
Affiliation(s)
- Yuye Shi
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yuhao Xue
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China
| | - Chunling Wang
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Liang Yu
- Department of Hematology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, People's Republic of China.,Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|
111
|
Zeng AGX, Bansal S, Jin L, Mitchell A, Chen WC, Abbas HA, Chan-Seng-Yue M, Voisin V, van Galen P, Tierens A, Cheok M, Preudhomme C, Dombret H, Daver N, Futreal PA, Minden MD, Kennedy JA, Wang JCY, Dick JE. A cellular hierarchy framework for understanding heterogeneity and predicting drug response in acute myeloid leukemia. Nat Med 2022; 28:1212-1223. [PMID: 35618837 DOI: 10.1038/s41591-022-01819-x] [Citation(s) in RCA: 151] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 04/07/2022] [Indexed: 02/08/2023]
Abstract
The treatment landscape of acute myeloid leukemia (AML) is evolving, with promising therapies entering clinical translation, yet patient responses remain heterogeneous, and biomarkers for tailoring treatment are lacking. To understand how disease heterogeneity links with therapy response, we determined the leukemia cell hierarchy makeup from bulk transcriptomes of more than 1,000 patients through deconvolution using single-cell reference profiles of leukemia stem, progenitor and mature cell types. Leukemia hierarchy composition was associated with functional, genomic and clinical properties and converged into four overall classes, spanning Primitive, Mature, GMP and Intermediate. Critically, variation in hierarchy composition along the Primitive versus GMP or Primitive versus Mature axes were associated with response to chemotherapy or drug sensitivity profiles of targeted therapies, respectively. A seven-gene biomarker derived from the Primitive versus Mature axis was associated with response to 105 investigational drugs. Cellular hierarchy composition constitutes a novel framework for understanding disease biology and advancing precision medicine in AML.
Collapse
Affiliation(s)
- Andy G X Zeng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Suraj Bansal
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Liqing Jin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Amanda Mitchell
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Weihsu Claire Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Biologics Discovery, Amgen British Columbia, Burnaby, BC, Canada
| | - Hussein A Abbas
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Peter van Galen
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Anne Tierens
- Laboratory Medicine Program, Hematopathology, University Health Network, Toronto, ON, Canada
| | - Meyling Cheok
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Claude Preudhomme
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Hervé Dombret
- Department of Hematology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Medical Oncology and Hematology, University Health Network, Toronto, ON, Canada
| | - James A Kennedy
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Division of Medical Oncology and Hematology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Medical Oncology and Hematology, University Health Network, Toronto, ON, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
112
|
Pianigiani G, Rocchio F, Peruzzi S, Andresen V, Bigerna B, Sorcini D, Capurro M, Gjertsen BT, Sportoletti P, Di Ianni M, Martelli MP, Brunetti L, Falini B. The absent/low expression of CD34 in NPM1-mutated AML is not related to cytoplasmic dislocation of NPM1 mutant protein. Leukemia 2022; 36:1931-1934. [PMID: 35568767 PMCID: PMC9252915 DOI: 10.1038/s41375-022-01593-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Giulia Pianigiani
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Francesca Rocchio
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy.,Research and Early Development, Dompé Farmaceutici S.p.A, Napoli, Italy
| | - Sara Peruzzi
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Vibeke Andresen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Barbara Bigerna
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Daniele Sorcini
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Michela Capurro
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medicine, Haematology Section, Haukeland University Hospital, Bergen, Norway
| | - Paolo Sportoletti
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Mauro Di Ianni
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Maria Paola Martelli
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy
| | - Lorenzo Brunetti
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy.,Department of Molecular and Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Brunangelo Falini
- Section of Hematology, Department of Medicine and Surgery, Center for Hemato-Oncological Research (CREO), University of Perugia, Perugia, Italy.
| |
Collapse
|
113
|
Umeda M, Ma J, Huang BJ, Hagiwara K, Westover T, Abdelhamed S, Barajas JM, Thomas ME, Walsh MP, Song G, Tian L, Liu Y, Chen X, Kolekar P, Tran Q, Foy SG, Maciaszek JL, Kleist AB, Leonti AR, Ju B, Easton J, Wu H, Valentine V, Valentine MB, Liu YC, Ries RE, Smith JL, Parganas E, Iacobucci I, Hiltenbrand R, Miller J, Myers JR, Rampersaud E, Rahbarinia D, Rusch M, Wu G, Inaba H, Wang YC, Alonzo TA, Downing JR, Mullighan CG, Pounds S, Babu MM, Zhang J, Rubnitz JE, Meshinchi S, Ma X, Klco JM. Integrated Genomic Analysis Identifies UBTF Tandem Duplications as a Recurrent Lesion in Pediatric Acute Myeloid Leukemia. Blood Cancer Discov 2022; 3:194-207. [PMID: 35176137 PMCID: PMC9780084 DOI: 10.1158/2643-3230.bcd-21-0160] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 08/27/2021] [Accepted: 01/24/2022] [Indexed: 01/21/2023] Open
Abstract
The genetics of relapsed pediatric acute myeloid leukemia (AML) has yet to be comprehensively defined. Here, we present the spectrum of genomic alterations in 136 relapsed pediatric AMLs. We identified recurrent exon 13 tandem duplications (TD) in upstream binding transcription factor (UBTF) in 9% of relapsed AML cases. UBTF-TD AMLs commonly have normal karyotype or trisomy 8 with cooccurring WT1 mutations or FLT3-ITD but not other known oncogenic fusions. These UBTF-TD events are stable during disease progression and are present in the founding clone. In addition, we observed that UBTF-TD AMLs account for approximately 4% of all de novo pediatric AMLs, are less common in adults, and are associated with poor outcomes and MRD positivity. Expression of UBTF-TD in primary hematopoietic cells is sufficient to enhance serial clonogenic activity and to drive a similar transcriptional program to UBTF-TD AMLs. Collectively, these clinical, genomic, and functional data establish UBTF-TD as a new recurrent mutation in AML. SIGNIFICANCE We defined the spectrum of mutations in relapsed pediatric AML and identified UBTF-TDs as a new recurrent genetic alteration. These duplications are more common in children and define a group of AMLs with intermediate-risk cytogenetic abnormalities, FLT3-ITD and WT1 alterations, and are associated with poor outcomes. See related commentary by Hasserjian and Nardi, p. 173. This article is highlighted in the In This Issue feature, p. 171.
Collapse
Affiliation(s)
- Masayuki Umeda
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jing Ma
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Benjamin J. Huang
- Department of Pediatrics, University of California, Benioff Children's Hospital, San Francisco, California
| | - Kohei Hagiwara
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Tamara Westover
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sherif Abdelhamed
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Juan M. Barajas
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Melvin E. Thomas
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michael P. Walsh
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Guangchun Song
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Liqing Tian
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yanling Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Xiaolong Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Pandurang Kolekar
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Quang Tran
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Scott G. Foy
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jamie L. Maciaszek
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Andrew B. Kleist
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Amanda R. Leonti
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Bengsheng Ju
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Huiyun Wu
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | | | - Yen-Chun Liu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Rhonda E. Ries
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jenny L. Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Evan Parganas
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ryan Hiltenbrand
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jonathan Miller
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jason R. Myers
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Evadnie Rampersaud
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Delaram Rahbarinia
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Todd A. Alonzo
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - James R. Downing
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - M. Madan Babu
- Department of Structural Biology and the Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jeffrey E. Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jeffery M. Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
114
|
Yoshino S, Suzuki HI. The molecular understanding of super-enhancer dysregulation in cancer. NAGOYA JOURNAL OF MEDICAL SCIENCE 2022; 84:216-229. [PMID: 35967935 PMCID: PMC9350580 DOI: 10.18999/nagjms.84.2.216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/20/2022] [Indexed: 11/30/2022]
Abstract
Abnormalities in the regulation of gene expression are associated with various pathological conditions. Among the distal regulatory elements in the genome, the activation of target genes by enhancers plays a central role in the formation of cell type-specific gene expression patterns. Super-enhancers are a subclass of enhancers that frequently contain multiple enhancer-like elements and are characterized by dense binding of master transcription factors and Mediator complexes and high signals of active histone marks. Super-enhancers have been studied in detail as important regulatory regions that control cell identity and contribute to the pathogenesis of diverse diseases. In cancer, super-enhancers have multifaceted roles by activating various oncogenes and other cancer-related genes and shaping characteristic gene expression patterns in cancer cells. Alterations in super-enhancer activities in cancer involve multiple mechanisms, including the dysregulation of transcription factors and the super-enhancer-associated genomic abnormalities. The study of super-enhancers could contribute to the identification of effective biomarkers and the development of cancer therapeutics targeting transcriptional addiction. In this review, we summarize the roles of super-enhancers in cancer biology, with a particular focus on hematopoietic malignancies, in which multiple super-enhancer alteration mechanisms have been reported.
Collapse
Affiliation(s)
- Seiko Yoshino
- Division of Molecular Oncology, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Hiroshi I. Suzuki
- Division of Molecular Oncology, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.
,Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| |
Collapse
|
115
|
IKAROS and MENIN coordinate therapeutically actionable leukemogenic gene expression in MLL-r acute myeloid leukemia. NATURE CANCER 2022; 3:595-613. [PMID: 35534777 PMCID: PMC9404532 DOI: 10.1038/s43018-022-00366-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 03/11/2022] [Indexed: 12/12/2022]
Abstract
Acute myeloid leukemia (AML) remains difficult to treat and requires new therapeutic approaches. Potent inhibitors of the chromatin-associated protein MENIN have recently entered human clinical trials, opening new therapeutic opportunities for some genetic subtypes of this disease. Using genome-scale functional genetic screens, we identified IKAROS (encoded by IKZF1) as an essential transcription factor in KMT2A (MLL1)-rearranged (MLL-r) AML that maintains leukemogenic gene expression while also repressing pathways for tumor suppression, immune regulation and cellular differentiation. Furthermore, IKAROS displays an unexpected functional cooperativity and extensive chromatin co-occupancy with mixed lineage leukemia (MLL)1-MENIN and the regulator MEIS1 and an extensive hematopoietic transcriptional complex involving homeobox (HOX)A10, MEIS1 and IKAROS. This dependency could be therapeutically exploited by inducing IKAROS protein degradation with immunomodulatory imide drugs (IMiDs). Finally, we demonstrate that combined IKAROS degradation and MENIN inhibition effectively disrupts leukemogenic transcriptional networks, resulting in synergistic killing of leukemia cells and providing a paradigm for improved drug targeting of transcription and an opportunity for rapid clinical translation.
Collapse
|
116
|
Jones LM, Starczynowski DT. IKAROS and MENIN in synergy in AML. NATURE CANCER 2022; 3:528-529. [PMID: 35624338 DOI: 10.1038/s43018-022-00387-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Affiliation(s)
- LaQuita M Jones
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- University of Cincinnati Cancer Center, Cincinnati, OH, USA
| | - Daniel T Starczynowski
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- University of Cincinnati Cancer Center, Cincinnati, OH, USA.
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
117
|
Escobar TM, Yu JR, Liu S, Lucero K, Vasilyev N, Nudler E, Reinberg D. Inheritance of repressed chromatin domains during S phase requires the histone chaperone NPM1. SCIENCE ADVANCES 2022; 8:eabm3945. [PMID: 35476441 PMCID: PMC9045712 DOI: 10.1126/sciadv.abm3945] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/09/2022] [Indexed: 06/14/2023]
Abstract
The epigenetic process safeguards cell identity during cell division through the inheritance of appropriate gene expression profiles. We demonstrated previously that parental nucleosomes are inherited by the same chromatin domains during DNA replication only in the case of repressed chromatin. We now show that this specificity is conveyed by NPM1, a histone H3/H4 chaperone. Proteomic analyses of late S-phase chromatin revealed NPM1 in association with both H3K27me3, an integral component of facultative heterochromatin, and MCM2, an integral component of the DNA replication machinery; moreover, NPM1 interacts directly with PRC2 and with MCM2. Given that NPM1 is essential, the inheritance of repressed chromatin domains was examined anew using mESCs expressing an auxin-degradable version of endogenous NPM1. Upon NPM1 degradation, cells accumulated in the G1-S phase of the cell cycle and parental nucleosome inheritance from repressed chromatin domains was markedly compromised. NPM1 chaperone activity may contribute to the integrity of this process as appropriate inheritance required the NPM1 acidic patches.
Collapse
Affiliation(s)
- Thelma M. Escobar
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Jia-Ray Yu
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Sanxiong Liu
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Kimberly Lucero
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Nikita Vasilyev
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Danny Reinberg
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
118
|
Cell Origin-Dependent Cooperativity of Mutant Dnmt3a and Npm1 in Clonal Hematopoiesis and Myeloid Malignancy. Blood Adv 2022; 6:3666-3677. [PMID: 35413095 DOI: 10.1182/bloodadvances.2022006968] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/31/2022] [Indexed: 11/20/2022] Open
Abstract
In adult acute myeloid leukemia (AML), acquisition of driver somatic mutations may be preceded by a benign state termed clonal hematopoiesis (CH). To develop therapeutic strategies to prevent leukemia development from CH, it is important to understand the mechanisms by which CH-driving and AML-driving mutations cooperate. Here, we use mice with inducible mutant alleles common in human CH (DNMT3AR882; mouse Dnmt3aR878H) and AML (NPM1c; mouse Npm1cA). We find that Dnmt3aR878H/+ hematopoietic stem cells (HSCs), but not multipotent progenitor cell (MPP) subsets, have reduced expression of cytokine and pro-inflammatory transcriptional signatures and a functional competitive advantage over their wild-type counterparts. Dnmt3aR878H/+ HSCs are the most potent cell type transformed by Npm1cA, generating myeloid malignancies in which few additional cooperating somatic mutation events were detected. At a molecular level, Npm1cA in cooperation with Dnmt3aR878H acutely increased accessibility of a distinct set of promoters in HSCs compared to MPP cells. These promoters were enriched for cell cycling, PI3K/AKT/mTOR signaling, stem cell signatures, and targets of transcription factors including NFAT and the chromatin binding factor HMGB1, which have been implicated in human AML. These results demonstrate cooperativity between pre-existing Dnmt3aR878H and Npm1cA at the chromatin level, where specific loci altered in accessibility by Npm1cA are dependent on cell context as well as Dnmt3a mutation status. These findings have implications for biological understanding and therapeutic intervention into transformation from CH to AML.
Collapse
|
119
|
Increasing Role of Targeted Immunotherapies in the Treatment of AML. Int J Mol Sci 2022; 23:ijms23063304. [PMID: 35328721 PMCID: PMC8953556 DOI: 10.3390/ijms23063304] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/11/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. The standard of care in medically and physically fit patients is intensive induction therapy. The majority of these intensively treated patients achieve a complete remission. However, a high number of these patients will experience relapse. In patients older than 60 years, the results are even worse. Therefore, new therapeutic approaches are desperately needed. One promising approach in high-risk leukemia to prevent relapse is the induction of the immune system simultaneously or after reduction of the initial tumor burden. Different immunotherapeutic approaches such as allogenic stem cell transplantation or donor lymphocyte infusions are already standard therapies, but other options for AML treatment are in the pipeline. Moreover, the therapeutic landscape in AML is rapidly changing, and in the last years, a number of immunogenic targets structures eligible for specific therapy, risk assessment or evaluation of disease course were determined. For example, leukemia-associated antigens (LAA) showed to be critical as biomarkers of disease state and survival, as well as markers of minimal residual disease (MRD). Yet many mechanisms and properties are still insufficiently understood, which also represents a great potential for this form of therapy. Therefore, targeted therapy as immunotherapy could turn into an efficient tool to clear residual disease, improve the outcome of AML patients and reduce the relapse risk. In this review, established but also emerging immunotherapeutic approaches for AML patients will be discussed.
Collapse
|
120
|
An oncogenic enhancer encodes selective selenium dependency in AML. Cell Stem Cell 2022; 29:386-399.e7. [PMID: 35108519 PMCID: PMC8903199 DOI: 10.1016/j.stem.2022.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 11/29/2021] [Accepted: 01/04/2022] [Indexed: 12/13/2022]
Abstract
Deregulation of transcription is a hallmark of acute myeloid leukemia (AML) that drives oncogenic expression programs and presents opportunities for therapeutic targeting. By integrating comprehensive pan-cancer enhancer landscapes with genetic dependency mapping, we find that AML-enriched enhancers encode for more selective tumor dependencies. We hypothesized that this approach could identify actionable dependencies downstream of oncogenic driver events and discovered a MYB-regulated AML-enriched enhancer regulating SEPHS2, a key component of the selenoprotein production pathway. Using a combination of patient samples and mouse models, we show that this enhancer upregulates SEPHS2, promoting selenoprotein production and antioxidant function required for AML survival. SEPHS2 and other selenoprotein pathway genes are required for AML growth in vitro. SEPHS2 knockout and selenium dietary restriction significantly delay leukemogenesis in vivo with little effect on normal hematopoiesis. These data validate the utility of enhancer mapping in target identification and suggest that selenoprotein production is an actionable target in AML.
Collapse
|
121
|
Xiao Q, Lei L, Ren J, Peng M, Jing Y, Jiang X, Huang J, Tao Y, Lin C, Yang J, Sun M, Tang L, Wei X, Yang Z, Zhang L. Mutant NPM1-Regulated FTO-Mediated m 6A Demethylation Promotes Leukemic Cell Survival via PDGFRB/ERK Signaling Axis. Front Oncol 2022; 12:817584. [PMID: 35211409 PMCID: PMC8862181 DOI: 10.3389/fonc.2022.817584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) with nucleophosmin 1 (NPM1) mutations exhibits distinct biological and clinical features, accounting for approximately one-third of AML. Recently, the N6-methyladenosine (m6A) RNA modification has emerged as a new epigenetic modification to contribute to tumorigenesis and development. However, there is limited knowledge on the role of m6A modifications in NPM1-mutated AML. In this study, the decreased m6A level was first detected and high expression of fat mass and obesity-associated protein (FTO) was responsible for the m6A suppression in NPM1-mutated AML. FTO upregulation was partially induced by NPM1 mutation type A (NPM1-mA) through impeding the proteasome pathway. Importantly, FTO promoted leukemic cell survival by facilitating cell cycle and inhibiting cell apoptosis. Mechanistic investigations demonstrated that FTO depended on its m6A RNA demethylase activity to activate PDGFRB/ERK signaling axis. Our findings indicate that FTO-mediated m6A demethylation plays an oncogenic role in NPM1-mutated AML and provide a new layer of epigenetic insight for future treatments of this distinctly leukemic entity.
Collapse
Affiliation(s)
- Qiaoling Xiao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Li Lei
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jun Ren
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Meixi Peng
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yipei Jing
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xueke Jiang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Junpeng Huang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yonghong Tao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Can Lin
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jing Yang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Minghui Sun
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Lisha Tang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xingyu Wei
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Zailin Yang
- Hematology Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Ling Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
122
|
Effective Menin inhibitor-based combinations against AML with MLL rearrangement or NPM1 mutation (NPM1c). Blood Cancer J 2022; 12:5. [PMID: 35017466 PMCID: PMC8752621 DOI: 10.1038/s41408-021-00603-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 12/16/2022] Open
Abstract
Treatment with Menin inhibitor (MI) disrupts the interaction between Menin and MLL1 or MLL1-fusion protein (FP), inhibits HOXA9/MEIS1, induces differentiation and loss of survival of AML harboring MLL1 re-arrangement (r) and FP, or expressing mutant (mt)-NPM1. Following MI treatment, although clinical responses are common, the majority of patients with AML with MLL1-r or mt-NPM1 succumb to their disease. Pre-clinical studies presented here demonstrate that genetic knockout or degradation of Menin or treatment with the MI SNDX-50469 reduces MLL1/MLL1-FP targets, associated with MI-induced differentiation and loss of viability. MI treatment also attenuates BCL2 and CDK6 levels. Co-treatment with SNDX-50469 and BCL2 inhibitor (venetoclax), or CDK6 inhibitor (abemaciclib) induces synergistic lethality in cell lines and patient-derived AML cells harboring MLL1-r or mtNPM1. Combined therapy with SNDX-5613 and venetoclax exerts superior in vivo efficacy in a cell line or PD AML cell xenografts harboring MLL1-r or mt-NPM1. Synergy with the MI-based combinations is preserved against MLL1-r AML cells expressing FLT3 mutation, also CRISPR-edited to introduce mtTP53. These findings highlight the promise of clinically testing these MI-based combinations against AML harboring MLL1-r or mtNPM1.
Collapse
|
123
|
Fiskus W, Daver N, Boettcher S, Mill CP, Sasaki K, Birdwell CE, Davis JA, Das K, Takahashi K, Kadia TM, DiNardo CD, Burrows F, Loghavi S, Khoury JD, Ebert BL, Bhalla KN. Activity of menin inhibitor ziftomenib (KO-539) as monotherapy or in combinations against AML cells with MLL1 rearrangement or mutant NPM1. Leukemia 2022; 36:2729-2733. [PMID: 36151141 PMCID: PMC9613474 DOI: 10.1038/s41375-022-01707-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/21/2022]
Affiliation(s)
- Warren Fiskus
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Naval Daver
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Steffen Boettcher
- grid.7400.30000 0004 1937 0650University of Zurich and University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Christopher P. Mill
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Koji Sasaki
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Christine E. Birdwell
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - John A. Davis
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Kaberi Das
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Koichi Takahashi
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Tapan M. Kadia
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Courtney D. DiNardo
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Francis Burrows
- grid.476498.00000 0004 6003 9775Kura Oncology, Inc., San Diego, CA 92130 USA
| | - Sanam Loghavi
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Joseph D. Khoury
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Benjamin L. Ebert
- grid.65499.370000 0001 2106 9910Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Boston, MA 02115 USA
| | - Kapil N. Bhalla
- grid.240145.60000 0001 2291 4776The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|
124
|
Mishra S, Liu J, Chai L, Tenen DG. Diverse functions of long noncoding RNAs in acute myeloid leukemia: emerging roles in pathophysiology, prognosis, and treatment resistance. Curr Opin Hematol 2022; 29:34-43. [PMID: 34854833 PMCID: PMC8647777 DOI: 10.1097/moh.0000000000000692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Advancements in the next-generation sequencing technologies have identified rare transcripts of long noncoding RNAs (lncRNAs) in the genome of cancers, including in acute myeloid leukemia (AML). The purpose of this review is to highlight the contribution of lncRNAs in AML pathogenesis, prognosis, and chemoresistance. RECENT FINDINGS Several studies have recently reported that deregulated lncRNAs are novel key players in the development of AML and are associated with AML pathophysiology and may serve as prognostic indicators. A few aberrantly expressed lncRNAs that correlated with the recurrent genetic mutations in AML such as NPM1 and RUNX1 have recently been characterized. Moreover, a few lncRNAs in MLL-rearranged leukemia have been described. Additionally, the involvement of lncRNAs in AML chemoresistance has been postulated. SUMMARY Investigating the functional roles of the noncoding regions including lncRNAs, may provide novel insights into the pathophysiology, refine the prognostic schema, and provide novel therapeutic treatment strategies in AML.
Collapse
Affiliation(s)
- Srishti Mishra
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Jun Liu
- Department of Pathology, Brigham & Women's Hospital
| | - Li Chai
- Department of Pathology, Brigham & Women's Hospital
| | - Daniel G Tenen
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
- Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
125
|
Novel NPM1 exon 5 mutations and gene fusions leading to aberrant cytoplasmic nucleophosmin in AML. Blood 2021; 138:2696-2701. [PMID: 34343258 PMCID: PMC9037756 DOI: 10.1182/blood.2021012732] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/17/2021] [Indexed: 11/20/2022] Open
Abstract
Nucleophosmin (NPM1) mutations in acute myeloid leukemia (AML) affect exon 12, but also sporadically affect exons 9 and 11, causing changes at the protein C-terminal end (tryptophan loss, nuclear export signal [NES] motif creation) that lead to aberrant cytoplasmic NPM1 (NPM1c+), detectable by immunohistochemistry. Combining immunohistochemistry and molecular analyses in 929 patients with AML, we found non-exon 12 NPM1 mutations in 5 (1.3%) of 387 NPM1c+ cases. Besides mutations in exons 9 (n = 1) and 11 (n = 1), novel exon 5 mutations were discovered (n = 3). Another exon 5 mutation was identified in an additional 141 patients with AML selected for wild-type NPM1 exon 12. Three NPM1 rearrangements (NPM1/RPP30, NPM1/SETBP1, NPM1/CCDC28A) were detected and characterized among 13 979 AML samples screened by cytogenetic/fluorescence in situ hybridization and RNA sequencing. Functional studies demonstrated that in AML cases, new NPM1 proteins harbored an efficient extra NES, either newly created or already present in the fusion partner, ensuring its cytoplasmic accumulation. Our findings support NPM1 cytoplasmic relocation as critical for leukemogenesis and reinforce the role of immunohistochemistry in predicting AML-associated NPM1 genetic lesions. This study highlights the need to develop new assays for molecular diagnosis and monitoring of NPM1-mutated AML.
Collapse
|
126
|
Zhao J, Xiu Y, Fu L, Dong Q, Borcherding N, Wang Y, Li Q, De Silva NS, Klein U, Boyce BF, Zhao C. TIFAB accelerates MLL-AF9-Induced acute myeloid leukemia through upregulation of HOXA9. iScience 2021; 24:103425. [PMID: 34877491 PMCID: PMC8633042 DOI: 10.1016/j.isci.2021.103425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
We previously showed stabilization of NIK-induced activation of NF-κB non-canonical signaling suppresses MLL-AF9-induced AML. In the current study, we demonstrate that deletion of NF-κB non-canonical RelB prevents the inhibitory effect of NIK stabilization in MLL-AF9 AML. Mechanistically, RelB suppresses its direct target, TIFAB, which is upregulated in human AML and correlates negatively with the survival of AML patients. Forced expression of TIFAB reverses NIK-induced impaired AML development through downregulation of RelB and upregulation of HOXA9. Consistent with upregulation of HOXA9, gene set enrichment analysis shows that forced expression of TIFAB blocks myeloid cell development, upregulates leukemia stem cell signature and induces similar gene expression patterns to those of HOXA9-MEIS1 and HOXA9-NUP98, and upregulates oxidative phosphorylation. Accordingly, forced expression of HOXA9 also largely releases the inhibitory impact of NIK stabilization via downregulation of RelB and upregulation of RelA. Our data suggest that NIK/RelB suppresses MLL-AF9-induced AML mainly through downregulation of TIFAB/HOXA9.
Collapse
Affiliation(s)
- Jinming Zhao
- Department of Pathology, Case Western Reserve University, Wolstein Research Building, Room 6503 2103 Cornell Road, Cleveland, OH 44106, USA.,Department of Pathology, China Medical University, 77 Puhe Road, Shenbei Xinqu, Shenyang, Liaoning Province, 110122, China
| | - Yan Xiu
- Department of Pathology, Case Western Reserve University, Wolstein Research Building, Room 6503 2103 Cornell Road, Cleveland, OH 44106, USA.,Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Lin Fu
- Department of Pathology, China Medical University, 77 Puhe Road, Shenbei Xinqu, Shenyang, Liaoning Province, 110122, China
| | - Qianze Dong
- Department of Pathology, Case Western Reserve University, Wolstein Research Building, Room 6503 2103 Cornell Road, Cleveland, OH 44106, USA.,Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Barnes-Jewish Hospital, Washington University in St Louis, MO 63110, USA
| | - Yang Wang
- Department of Pathology, Case Western Reserve University, Wolstein Research Building, Room 6503 2103 Cornell Road, Cleveland, OH 44106, USA
| | - Qingchang Li
- Department of Pathology, China Medical University, 77 Puhe Road, Shenbei Xinqu, Shenyang, Liaoning Province, 110122, China
| | | | - Ulf Klein
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James, Leeds LS9 7TF, UK
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Chen Zhao
- Department of Pathology, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106, USA.,Department of Pathology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA.,Department of Pathology, Case Western Reserve University, Wolstein Research Building, Room 6523 2103 Cornell Road, Cleveland, OH 44106, USA
| |
Collapse
|
127
|
Immunosuppression and outcomes in adult patients with de novo acute myeloid leukemia with normal karyotypes. Proc Natl Acad Sci U S A 2021; 118:2116427118. [PMID: 34845035 PMCID: PMC8673586 DOI: 10.1073/pnas.2116427118] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 11/18/2022] Open
Abstract
Acute myeloid leukemia (AML) patients rarely have long first remissions (LFRs; >5 y) after standard-of-care chemotherapy, unless classified as favorable risk at presentation. Identification of the mechanisms responsible for long vs. more typical, standard remissions may help to define prognostic determinants for chemotherapy responses. Using exome sequencing, RNA-sequencing, and functional immunologic studies, we characterized 28 normal karyotype (NK)-AML patients with >5 y first remissions after chemotherapy (LFRs) and compared them to a well-matched group of 31 NK-AML patients who relapsed within 2 y (standard first remissions [SFRs]). Our combined analyses indicated that genetic-risk profiling at presentation (as defined by European LeukemiaNet [ELN] 2017 criteria) was not sufficient to explain the outcomes of many SFR cases. Single-cell RNA-sequencing studies of 15 AML samples showed that SFR AML cells differentially expressed many genes associated with immune suppression. The bone marrow of SFR cases had significantly fewer CD4+ Th1 cells; these T cells expressed an exhaustion signature and were resistant to activation by T cell receptor stimulation in the presence of autologous AML cells. T cell activation could be restored by removing the AML cells or blocking the inhibitory major histocompatibility complex class II receptor, LAG3. Most LFR cases did not display these features, suggesting that their AML cells were not as immunosuppressive. These findings were confirmed and extended in an independent set of 50 AML cases representing all ELN 2017 risk groups. AML cell-mediated suppression of CD4+ T cell activation at presentation is strongly associated with unfavorable outcomes in AML patients treated with standard chemotherapy.
Collapse
|
128
|
Wu HC, Rérolle D, Berthier C, Hleihel R, Sakamoto T, Quentin S, Benhenda S, Morganti C, Wu C, Conte L, Rimsky S, Sebert M, Clappier E, Souquere S, Gachet S, Soulier J, Durand S, Trowbridge JJ, Bénit P, Rustin P, El Hajj H, Raffoux E, Ades L, Itzykson R, Dombret H, Fenaux P, Espeli O, Kroemer G, Brunetti L, Mak TW, Lallemand-Breitenbach V, Bazarbachi A, Falini B, Ito K, Martelli MP, de Thé H. Actinomycin D Targets NPM1c-Primed Mitochondria to Restore PML-Driven Senescence in AML Therapy. Cancer Discov 2021; 11:3198-3213. [PMID: 34301789 PMCID: PMC7612574 DOI: 10.1158/2159-8290.cd-21-0177] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/07/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) pathogenesis often involves a mutation in the NPM1 nucleolar chaperone, but the bases for its transforming properties and overall association with favorable therapeutic responses remain incompletely understood. Here we demonstrate that an oncogenic mutant form of NPM1 (NPM1c) impairs mitochondrial function. NPM1c also hampers formation of promyelocytic leukemia (PML) nuclear bodies (NB), which are regulators of mitochondrial fitness and key senescence effectors. Actinomycin D (ActD), an antibiotic with unambiguous clinical efficacy in relapsed/refractory NPM1c-AMLs, targets these primed mitochondria, releasing mitochondrial DNA, activating cyclic GMP-AMP synthase signaling, and boosting reactive oxygen species (ROS) production. The latter restore PML NB formation to drive TP53 activation and senescence of NPM1c-AML cells. In several models, dual targeting of mitochondria by venetoclax and ActD synergized to clear AML and prolong survival through targeting of PML. Our studies reveal an unexpected role for mitochondria downstream of NPM1c and implicate a mitochondrial/ROS/PML/TP53 senescence pathway as an effector of ActD-based therapies. SIGNIFICANCE ActD induces complete remissions in NPM1-mutant AMLs. We found that NPM1c affects mitochondrial biogenesis and PML NBs. ActD targets mitochondria, yielding ROS which enforce PML NB biogenesis and restore senescence. Dual targeting of mitochondria with ActD and venetoclax sharply potentiates their anti-AML activities in vivo. This article is highlighted in the In This Issue feature, p. 2945.
Collapse
Affiliation(s)
- Hsin-Chieh Wu
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Domitille Rérolle
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Caroline Berthier
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Rita Hleihel
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Internal Medicine and Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
- Department of Experimental Pathology, Microbiology and Immunology, American University of Beirut, Beirut, Lebanon
| | - Takashi Sakamoto
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Samuel Quentin
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Shirine Benhenda
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research and Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Chengchen Wu
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Lidio Conte
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli, ” Napoli, Italy
| | - Sylvie Rimsky
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
| | - Marie Sebert
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Emmanuelle Clappier
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Sylvie Souquere
- Institut Gustave Roussy, Cell Biology and Metabolomics Platforms, INSERM UMS 3655, Villejuif, France
| | - Stéphanie Gachet
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Jean Soulier
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Sylvère Durand
- Institut Gustave Roussy, Cell Biology and Metabolomics Platforms, INSERM UMS 3655, Villejuif, France
| | | | - Paule Bénit
- INSERM, U1141 Hôpital Robert Debré, Paris France
| | | | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, American University of Beirut, Beirut, Lebanon
| | - Emmanuel Raffoux
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Lionel Ades
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Raphael Itzykson
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Hervé Dombret
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Pierre Fenaux
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| | - Olivier Espeli
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
| | - Guido Kroemer
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli, ” Napoli, Italy
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Lorenzo Brunetti
- Hematology, Department of Medicine and surgery, University of Perugia, Perugia, Italy
| | - Tak W. Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Valérie Lallemand-Breitenbach
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
| | - Ali Bazarbachi
- Department of Internal Medicine and Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Brunangelo Falini
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research and Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, Bronx, New York
| | | | - Hugues de Thé
- Collège de France, Oncologie Cellulaire et Moléculaire, PSL University, INSERM UMR 1050, CNRS UMR 7241, Paris, France
- Université de Paris, INSERM U944, CNRS UMR 7212, IRSL, Hôpital St. Louis, Paris, France
- Department of Hematology, Hôpital Saint Louis (Assistance publique Hôpitaux de Paris) and Paris University, Paris, France
| |
Collapse
|
129
|
Alayed K, Meyerson HJ. Decreased CD177 pos neutrophils in myeloid neoplasms is associated with NPM1, RUNX1, TET2, and U2AF1 S34F mutations. Leuk Res 2021; 112:106752. [PMID: 34896936 DOI: 10.1016/j.leukres.2021.106752] [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: 07/20/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 11/26/2022]
Abstract
A decreased percentage of CD177pos neutrophils is frequently present in MDS and AML and is a useful flow cytometry (FCM) marker for the identification of MDS. The underlying mechanism leading to the low percentage of CD177pos neutrophils in MDS has not been explained. The aim of this study was to identify whether specific somatic mutations in myeloid neoplasms are associated with the low percentage of CD177pos neutrophils. 507 myeloid neoplasms with one or more pathogenic molecular abnormality identified by NGS and in which CD177 expression was assessed were evaluated. Correlation with CD177 expression was determined for 39 variables (including genes mutated, diagnostic groups and gender) using a 40 % cutoff level for low CD177 expression. In multivariate analysis mutations involving NPM1 (OD 0.26), RUNX1 (OD 0.39), TET2 (OD 0.58), and U2AF1 S34F (OD 0.25) were associated with low percentage of CD177pos neutrophils when all cases were evaluated. JAK2 (OD 2.5) alteration was associated with increased percentage of CD177pos neutrophils. Differences were noted between diagnostic subgroups with no single mutation associated with decreased CD177pos neutrophils in MDS and CCUS. The findings demonstrate an association between the percentage of CD177pos neutrophils and somatically acquired mutations involving several genes.
Collapse
Affiliation(s)
- Khaled Alayed
- King Saud University Medical City, Riyadh, Saudi Arabia
| | - Howard J Meyerson
- University Hospitals Cleveland Medical Center/ Case Western Reserve University, Cleveland, OH, United States.
| |
Collapse
|
130
|
Djamai H, Berrou J, Dupont M, Coudé MM, Delord M, Clappier E, Marceau-Renaut A, Kaci A, Raffoux E, Itzykson R, Berthier C, Wu HC, Hleihel R, Bazarbachi A, de Thé H, Baruchel A, Gardin C, Dombret H, Braun T. Biological Effects of BET Inhibition by OTX015 (MK-8628) and JQ1 in NPM1-Mutated (NPM1c) Acute Myeloid Leukemia (AML). Biomedicines 2021; 9:biomedicines9111704. [PMID: 34829934 PMCID: PMC8615962 DOI: 10.3390/biomedicines9111704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
BET inhibitors (BETi) including OTX015 (MK-8628) and JQ1 demonstrated antileukemic activity including NPM1c AML cells. Nevertheless, the biological consequences of BETi in NPM1c AML were not fully investigated. Even if of better prognosis AML patients with NPM1c may relapse and treatment remains difficult. Differentiation-based therapy by all trans retinoic acid (ATRA) combined with arsenic trioxide (ATO) demonstrated activity in NPM1c AML. We found that BETi, similar to ATO + ATRA, induced differentiation and apoptosis which was TP53 independent in the NPM1c cell line OCI-AML3 and primary cells. Furthermore, BETi induced proteasome-dependent degradation of NPM1c. BETi degraded NPM1c in the cytosol while BRD4 is degraded in the nucleus which suggests that restoration of the NPM1/BRD4 equilibrium in the nucleus of NPM1c cells is essential for the efficacy of BETi. While ATO + ATRA had significant biological activity in NPM1c IMS-M2 cell line, those cells were resistant to BETi. Gene profiling revealed that IMS-M2 cells probably resist to BETi by upregulation of LSC pathways independently of the downregulation of a core BET-responsive transcriptional program. ATO + ATRA downregulated a NPM1c specific HOX gene signature while anti-leukemic effects of BETi appear HOX gene independent. Our preclinical results encourage clinical testing of BETi in NPM1c AML patients.
Collapse
Affiliation(s)
- Hanane Djamai
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
| | - Jeannig Berrou
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
| | - Mélanie Dupont
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
| | - Marie-Magdelaine Coudé
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
- Laboratory of Hematology, Hôpital Saint-Louis, AP-HP, Université de Paris, 75010 Paris, France;
| | - Marc Delord
- Bioinformatics, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France;
| | - Emmanuelle Clappier
- Laboratory of Hematology, Hôpital Saint-Louis, AP-HP, Université de Paris, 75010 Paris, France;
| | | | - Anna Kaci
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
| | - Emmanuel Raffoux
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
- Leukemia Unit, Hematology Department, Hôpital Saint-Louis, AP-HP, Université de Paris, 75010 Paris, France;
| | - Raphaël Itzykson
- Leukemia Unit, Hematology Department, Hôpital Saint-Louis, AP-HP, Université de Paris, 75010 Paris, France;
- INSERM U944—CNRS UMR7212, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (C.B.); (H.-C.W.); (H.d.T.)
| | - Caroline Berthier
- INSERM U944—CNRS UMR7212, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (C.B.); (H.-C.W.); (H.d.T.)
| | - Hsin-Chieh Wu
- INSERM U944—CNRS UMR7212, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (C.B.); (H.-C.W.); (H.d.T.)
| | - Rita Hleihel
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 113-6044, Lebanon; (R.H.); (A.B.)
| | - Ali Bazarbachi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 113-6044, Lebanon; (R.H.); (A.B.)
| | - Hugues de Thé
- INSERM U944—CNRS UMR7212, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (C.B.); (H.-C.W.); (H.d.T.)
| | - André Baruchel
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
- Department of Pediatric Hemato-Immunology, Hôpital Robert Debré, AP-HP, Université de Paris, 75010 Paris, France
| | - Claude Gardin
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
- Hematology Department, Hôpital Avicenne, AP-HP, Université de Paris, 93000 Bobigny, France
| | - Hervé Dombret
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
- Leukemia Unit, Hematology Department, Hôpital Saint-Louis, AP-HP, Université de Paris, 75010 Paris, France;
| | - Thorsten Braun
- Laboratoire de Transfert des Leucémies, URP-3518, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France; (H.D.); (J.B.); (M.D.); (M.-M.C.); (A.K.); (E.R.); (A.B.); (C.G.); (H.D.)
- Hematology Department, Hôpital Avicenne, AP-HP, Université de Paris, 93000 Bobigny, France
- Correspondence: ; Tel.: +33-148957072
| |
Collapse
|
131
|
Peng M, Ren J, Jing Y, Jiang X, Xiao Q, Huang J, Tao Y, Lei L, Wang X, Yang Z, Yang Z, Zhan Q, Lin C, Jin G, Zhang X, Zhang L. Tumour-derived small extracellular vesicles suppress CD8+ T cell immune function by inhibiting SLC6A8-mediated creatine import in NPM1-mutated acute myeloid leukaemia. J Extracell Vesicles 2021; 10:e12168. [PMID: 34807526 PMCID: PMC8607980 DOI: 10.1002/jev2.12168] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/11/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukaemia (AML) carrying nucleophosmin (NPM1) mutations has been defined as a distinct entity of acute leukaemia. Despite remarkable improvements in diagnosis and treatment, the long-term outcomes for this entity remain unsatisfactory. Emerging evidence suggests that leukaemia, similar to other malignant diseases, employs various mechanisms to evade killing by immune cells. However, the mechanism of immune escape in NPM1-mutated AML remains unknown. In this study, both serum and leukemic cells from patients with NPM1-mutated AML impaired the immune function of CD8+ T cells in a co-culture system. Mechanistically, leukemic cells secreted miR-19a-3p into the tumour microenvironment (TME) via small extracellular vesicles (sEVs), which was controlled by the NPM1-mutated protein/CCCTC-binding factor (CTCF)/poly (A)-binding protein cytoplasmic 1 (PABPC1) signalling axis. sEV-related miR-19a-3p was internalized by CD8+ T cells and directly repressed the expression of solute-carrier family 6 member 8 (SLC6A8; a creatine-specific transporter) to inhibit creatine import. Decreased creatine levels can reduce ATP production and impair CD8+ T cell immune function, leading to immune escape by leukemic cells. In summary, leukemic cell-derived sEV-related miR-19a-3p confers immunosuppression to CD8+ T cells by targeting SLC6A8-mediated creatine import, indicating that sEV-related miR-19a-3p might be a promising therapeutic target for NPM1-mutated AML.
Collapse
Affiliation(s)
- Meixi Peng
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Jun Ren
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Yipei Jing
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Xueke Jiang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Qiaoling Xiao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Junpeng Huang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Yonghong Tao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Li Lei
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Xin Wang
- Department of HematologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zailin Yang
- Department of Clinical Laboratory The Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing University Cancer HospitalChongqingChina
| | - Zesong Yang
- Department of HematologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Qian Zhan
- The Center for Clinical Molecular Medical detectionThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Can Lin
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Guoxiang Jin
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Xian Zhang
- Immunology ProgramMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Ling Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| |
Collapse
|
132
|
Blasi F, Bruckmann C. MEIS1 in Hematopoiesis and Cancer. How MEIS1-PBX Interaction Can Be Used in Therapy. J Dev Biol 2021; 9:jdb9040044. [PMID: 34698191 PMCID: PMC8544432 DOI: 10.3390/jdb9040044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 11/26/2022] Open
Abstract
Recently MEIS1 emerged as a major determinant of the MLL-r leukemic phenotype. The latest and most efficient drugs effectively decrease the levels of MEIS1 in cancer cells. Together with an overview of the latest drugs developed to target MEIS1 in MLL-r leukemia, we review, in detail, the role of MEIS1 in embryonic and adult hematopoiesis and suggest how a more profound knowledge of MEIS1 biochemistry can be used to design potent and effective drugs against MLL-r leukemia. In addition, we present data showing that the interaction between MEIS1 and PBX1 can be blocked efficiently and might represent a new avenue in anti-MLL-r and anti-leukemic therapy.
Collapse
|
133
|
Modeling IKZF1 lesions in B-ALL reveals distinct chemosensitivity patterns and potential therapeutic vulnerabilities. Blood Adv 2021; 5:3876-3890. [PMID: 34492683 DOI: 10.1182/bloodadvances.2020002408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 05/26/2021] [Indexed: 12/25/2022] Open
Abstract
IKAROS family zinc finger 1 (IKZF1) alterations represent a diverse group of genetic lesions that are associated with an increased risk of relapse in B-cell acute lymphoblastic leukemia. Due to the heterogeneity of concomitant lesions, it remains unclear how IKZF1 abnormalities directly affect cell function and therapy resistance, and whether their consideration as a prognostic indicator is valuable in improving outcome. CRISPR/Cas9 strategies were used to engineer multiple panels of isogeneic lymphoid leukemia cell lines with a spectrum of IKZF1 lesions to measure changes in chemosensitivity, gene expression, cell cycle, and in vivo engraftment that can be linked to loss of IKAROS protein. IKZF1 knockout and heterozygous null cells displayed relative resistance to a number of common therapies for B-cell acute lymphoblastic leukemia, including dexamethasone, asparaginase, and daunorubicin. Transcription profiling revealed a stem/myeloid cell-like phenotype and JAK/STAT upregulation after IKAROS loss. A CRISPR homology-directed repair strategy was also used to knock-in the dominant-negative IK6 isoform into the endogenous locus, and a similar drug resistance profile, with the exception of retained dexamethasone sensitivity, was observed. Interestingly, IKZF1 knockout and IK6 knock-in cells both have significantly increased sensitivity to cytarabine, likely owing to marked downregulation of SAMHD1 after IKZF1 knockout. Both types of IKZF1 lesions decreased the survival time of xenograft mice, with higher numbers of circulating blasts and increased organ infiltration. Given these findings, exact specification of IKZF1 status in patients may be a beneficial addition to risk stratification and could inform therapy.
Collapse
|
134
|
Lu B, Zou C, Yang M, He Y, He J, Zhang C, Chen S, Yu J, Liu KY, Cao Q, Zhao W. Pharmacological Inhibition of Core Regulatory Circuitry Liquid-liquid Phase Separation Suppresses Metastasis and Chemoresistance in Osteosarcoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101895. [PMID: 34432948 PMCID: PMC8529446 DOI: 10.1002/advs.202101895] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/05/2021] [Indexed: 05/29/2023]
Abstract
Liquid-liquid phase-separated (LLPS) transcriptional factor assemblies at super-enhancers (SEs) provide a conceptual framework for underlying transcriptional control in mammal cells. However, the mechanistic understanding of LLPS in aberrant transcription driven by dysregulation of SEs in human malignancies is still elusive. By integrating SE profiling and core regulatory circuitry (CRC) calling algorithm, the CRC of metastatic and chemo-resistant osteosarcoma is delineated. CRC components, HOXB8 and FOSL1, produce dense and dynamic phase-separated droplets in vitro and liquid-like puncta in cell nuclei. Disruption of CRC phase separation decreases the chromatin accessibility in SE regions and inhibits the release of RNA polymerase II from the promoter of SE-driven genes. Importantly, absence of CRC key component causes a reduction in osteosarcoma tumor growth and metastasis. Moreover, it is shown that CRC condensates can be specifically attenuated by the H3K27 demethylase inhibitor, GSK-J4. Pharmacological inhibition of the CRC phase separation results in metastasis suppression and re-sensitivity to chemotherapy drugs in patient-derived xenograft model. Taken together, this study reveals a previously unknown mechanism that CRC factors formed LLPS condensates, and provides a phase separation-based pharmacological strategy to target undruggable CRC components for the treatment of metastatic and chemo-resistant osteosarcoma.
Collapse
Affiliation(s)
- Bing Lu
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Changye Zou
- Musculoskeletal Oncology CenterThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhou510080China
| | - Meiling Yang
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Yangyang He
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Jincan He
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Chuanxia Zhang
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Siyun Chen
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Jiaming Yu
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Kilia Yun Liu
- Department of UrologyNorthwestern University Feinberg School of MedicineChicagoIL60611USA
| | - Qi Cao
- Department of UrologyNorthwestern University Feinberg School of MedicineChicagoIL60611USA
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern University Feinberg School of MedicineChicagoIL60611USA
| | - Wei Zhao
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| |
Collapse
|
135
|
Hindley A, Catherwood MA, McMullin MF, Mills KI. Significance of NPM1 Gene Mutations in AML. Int J Mol Sci 2021; 22:ijms221810040. [PMID: 34576201 PMCID: PMC8467861 DOI: 10.3390/ijms221810040] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/16/2022] Open
Abstract
The aim of this literature review is to examine the significance of the nucleophosmin 1 (NPM1) gene in acute myeloid leukaemia (AML). This will include analysis of the structure and normal cellular function of NPM1, the type of mutations commonly witnessed in NPM1, and the mechanism by which this influences the development and progression of AML. The importance of NPM1 mutation on prognosis and the treatment options available to patients will also be reviewed along with current guidelines recommending the rapid return of NPM1 mutational screening results and the importance of employing a suitable laboratory assay to achieve this. Finally, future developments in the field including research into new therapies targeting NPM1 mutated AML are considered.
Collapse
Affiliation(s)
- Andrew Hindley
- Clinical Haematology, Belfast City Hospital, Belfast BT9 7AB, UK;
- Correspondence:
| | | | - Mary Frances McMullin
- Centre for Medical Education, Queen’s University Belfast, Belfast BT7 1NN, UK;
- Northern Ireland and Belfast Health and Social Care Trust, Belfast BT9 7AB, UK
| | - Ken I. Mills
- Patrick G Johnston Center for Cancer Research, Queens University Belfast, Belfast BT9 7AE, UK;
| |
Collapse
|
136
|
Venanzi A, Rossi R, Martino G, Annibali O, Avvisati G, Mameli MG, Sportoletti P, Tiacci E, Falini B, Martelli MP. A Curious Novel Combination of Nucleophosmin ( NPM1) Gene Mutations Leading to Aberrant Cytoplasmic Dislocation of NPM1 in Acute Myeloid Leukemia (AML). Genes (Basel) 2021; 12:genes12091426. [PMID: 34573408 PMCID: PMC8468273 DOI: 10.3390/genes12091426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 11/16/2022] Open
Abstract
Nucleophosmin (NPM1) mutations occurring in acute myeloid leukemia (AML) (about 50 so far identified) cluster almost exclusively in exon 12 and lead to common changes at the NPM1 mutants C-terminus, i.e., loss of tryptophans 288 and 290 (or 290 alone) and creation of a new nuclear export signal (NES), at the bases of exportin-1(XPO1)-mediated aberrant cytoplasmic NPM1. Immunohistochemistry (IHC) detects cytoplasmic NPM1 and is predictive of the molecular alteration. Besides IHC and molecular sequencing, Western blotting (WB) with anti-NPM1 mutant specific antibodies is another approach to identify NPM1-mutated AML. Here, we show that among 382 AML cases with NPM1 exon 12 mutations, one was not recognized by WB, and describe the discovery of a novel combination of two mutations involving exon 12. This appeared as a conventional mutation A with the known TCTG nucleotides insertion/duplication accompanied by a second event (i.e., an 8-nucleotide deletion occurring 15 nucleotides downstream of the TCTG insertion), resulting in a new C-terminal protein sequence. Strikingly, the sequence included a functional NES ensuring cytoplasmic relocation of the new mutant supporting the role of cytoplasmic NPM1 as critical in AML leukemogenesis.
Collapse
Affiliation(s)
- Alessandra Venanzi
- Hematology and Clinical Immunology, Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, 06132 Perugia, Italy; (A.V.); (R.R.); (P.S.); (E.T.); (B.F.)
- Hematology Section, “Santa Maria della Misericordia” Hospital of Perugia, 06132 Perugia, Italy;
| | - Roberta Rossi
- Hematology and Clinical Immunology, Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, 06132 Perugia, Italy; (A.V.); (R.R.); (P.S.); (E.T.); (B.F.)
| | - Giovanni Martino
- Pathology Unit, Azienda Ospedaliera Santa Maria di Terni, University of Perugia, 05100 Terni, Italy;
- Department of Pathology, AOU Cagliari, University of Cagliari, 09042 Cagliari, Italy
| | - Ombretta Annibali
- Hematology and Stem Cell Transplant Unit, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (O.A.); (G.A.)
| | - Giuseppe Avvisati
- Hematology and Stem Cell Transplant Unit, Campus Bio-Medico University of Rome, 00128 Rome, Italy; (O.A.); (G.A.)
| | - Maria Grazia Mameli
- Hematology Section, “Santa Maria della Misericordia” Hospital of Perugia, 06132 Perugia, Italy;
| | - Paolo Sportoletti
- Hematology and Clinical Immunology, Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, 06132 Perugia, Italy; (A.V.); (R.R.); (P.S.); (E.T.); (B.F.)
- Hematology Section, “Santa Maria della Misericordia” Hospital of Perugia, 06132 Perugia, Italy;
| | - Enrico Tiacci
- Hematology and Clinical Immunology, Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, 06132 Perugia, Italy; (A.V.); (R.R.); (P.S.); (E.T.); (B.F.)
- Hematology Section, “Santa Maria della Misericordia” Hospital of Perugia, 06132 Perugia, Italy;
| | - Brunangelo Falini
- Hematology and Clinical Immunology, Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, 06132 Perugia, Italy; (A.V.); (R.R.); (P.S.); (E.T.); (B.F.)
- Hematology Section, “Santa Maria della Misericordia” Hospital of Perugia, 06132 Perugia, Italy;
| | - Maria Paola Martelli
- Hematology and Clinical Immunology, Centro di Ricerche Emato-Oncologiche (CREO), University of Perugia, 06132 Perugia, Italy; (A.V.); (R.R.); (P.S.); (E.T.); (B.F.)
- Hematology Section, “Santa Maria della Misericordia” Hospital of Perugia, 06132 Perugia, Italy;
- Correspondence:
| |
Collapse
|
137
|
Abstract
Herculean efforts by the Wellcome Sanger Institute, the National Cancer Institute, and the National Human Genome Research Institute to sequence thousands of tumors representing all major cancer types have yielded more than 700 genes that contribute to neoplastic growth when mutated, amplified, or deleted. While some of these genes (now included in the COSMIC Cancer Gene Census) encode proteins previously identified in hypothesis-driven experiments (oncogenic transcription factors, protein kinases, etc.), additional classes of cancer drivers have emerged, perhaps none more surprisingly than RNA-binding proteins (RBPs). Over 40 RBPs responsible for virtually all aspects of RNA metabolism, from synthesis to degradation, are recurrently mutated in cancer, and just over a dozen are considered major cancer drivers. This Review investigates whether and how their RNA-binding activities pertain to their oncogenic functions. Focusing on several well-characterized steps in RNA metabolism, we demonstrate that for virtually all cancer-driving RBPs, RNA processing activities are either abolished (the loss-of-function phenotype) or carried out with low fidelity (the LoFi phenotype). Conceptually, this suggests that in normal cells, RBPs act as gatekeepers maintaining proper RNA metabolism and the "balanced" proteome. From the practical standpoint, at least some LoFi phenotypes create therapeutic vulnerabilities, which are beginning to be exploited in the clinic.
Collapse
|
138
|
Cao Z, Budinich KA, Huang H, Ren D, Lu B, Zhang Z, Chen Q, Zhou Y, Huang YH, Alikarami F, Kingsley MC, Lenard AK, Wakabayashi A, Khandros E, Bailis W, Qi J, Carroll MP, Blobel GA, Faryabi RB, Bernt KM, Berger SL, Shi J. ZMYND8-regulated IRF8 transcription axis is an acute myeloid leukemia dependency. Mol Cell 2021; 81:3604-3622.e10. [PMID: 34358447 PMCID: PMC8932643 DOI: 10.1016/j.molcel.2021.07.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023]
Abstract
The transformed state in acute leukemia requires gene regulatory programs involving transcription factors and chromatin modulators. Here, we uncover an IRF8-MEF2D transcriptional circuit as an acute myeloid leukemia (AML)-biased dependency. We discover and characterize the mechanism by which the chromatin "reader" ZMYND8 directly activates IRF8 in parallel with the MYC proto-oncogene through their lineage-specific enhancers. ZMYND8 is essential for AML proliferation in vitro and in vivo and associates with MYC and IRF8 enhancer elements that we define in cell lines and in patient samples. ZMYND8 occupancy at IRF8 and MYC enhancers requires BRD4, a transcription coactivator also necessary for AML proliferation. We show that ZMYND8 binds to the ET domain of BRD4 via its chromatin reader cassette, which in turn is required for proper chromatin occupancy and maintenance of leukemic growth in vivo. Our results rationalize ZMYND8 as a potential therapeutic target for modulating essential transcriptional programs in AML.
Collapse
Affiliation(s)
- Zhendong Cao
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Krista A Budinich
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hua Huang
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Diqiu Ren
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bin Lu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Zhen Zhang
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qingzhou Chen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yeqiao Zhou
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yu-Han Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Fatemeh Alikarami
- Division of Pediatric Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Molly C Kingsley
- Division of Pediatric Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandra K Lenard
- Division of Pediatric Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Aoi Wakabayashi
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eugene Khandros
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Will Bailis
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Martin P Carroll
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gerd A Blobel
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Robert B Faryabi
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathrin M Bernt
- Division of Pediatric Oncology, Department of Pediatrics, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shelley L Berger
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junwei Shi
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
139
|
Hassan JJ, Lieske A, Dörpmund N, Klatt D, Hoffmann D, Kleppa MJ, Kustikova OS, Stahlhut M, Schwarzer A, Schambach A, Maetzig T. A Multiplex CRISPR-Screen Identifies PLA2G4A as Prognostic Marker and Druggable Target for HOXA9 and MEIS1 Dependent AML. Int J Mol Sci 2021; 22:ijms22179411. [PMID: 34502319 PMCID: PMC8431012 DOI: 10.3390/ijms22179411] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 12/03/2022] Open
Abstract
HOXA9 and MEIS1 are frequently upregulated in acute myeloid leukemia (AML), including those with MLL-rearrangement. Because of their pivotal role in hemostasis, HOXA9 and MEIS1 appear non-druggable. We, thus, interrogated gene expression data of pre-leukemic (overexpressing Hoxa9) and leukemogenic (overexpressing Hoxa9 and Meis1; H9M) murine cell lines to identify cancer vulnerabilities. Through gene expression analysis and gene set enrichment analyses, we compiled a list of 15 candidates for functional validation. Using a novel lentiviral multiplexing approach, we selected and tested highly active sgRNAs to knockout candidate genes by CRISPR/Cas9, and subsequently identified a H9M cell growth dependency on the cytosolic phospholipase A2 (PLA2G4A). Similar results were obtained by shRNA-mediated suppression of Pla2g4a. Remarkably, pharmacologic inhibition of PLA2G4A with arachidonyl trifluoromethyl ketone (AACOCF3) accelerated the loss of H9M cells in bulk cultures. Additionally, AACOCF3 treatment of H9M cells reduced colony numbers and colony sizes in methylcellulose. Moreover, AACOCF3 was highly active in human AML with MLL rearrangement, in which PLA2G4A was significantly higher expressed than in AML patients without MLL rearrangement, and is sufficient as an independent prognostic marker. Our work, thus, identifies PLA2G4A as a prognostic marker and potential therapeutic target for H9M-dependent AML with MLL-rearrangement.
Collapse
MESH Headings
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- CRISPR-Cas Systems
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Group IV Phospholipases A2/antagonists & inhibitors
- Group IV Phospholipases A2/genetics
- High-Throughput Screening Assays
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Myeloid Ecotropic Viral Integration Site 1 Protein/genetics
- Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Jacob Jalil Hassan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
| | - Anna Lieske
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
- Department of Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Nicole Dörpmund
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
- Department of Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Denise Klatt
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
| | - Dirk Hoffmann
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
| | - Marc-Jens Kleppa
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
| | - Olga S. Kustikova
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
| | - Maike Stahlhut
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
| | - Adrian Schwarzer
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tobias Maetzig
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (J.J.H.); (A.L.); (N.D.); (D.K.); (D.H.); (M.-J.K.); (O.S.K.); (M.S.); (A.S.); (A.S.)
- Department of Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
- Correspondence:
| |
Collapse
|
140
|
NPM1 Mutational Status Underlines Different Biological Features in Pediatric AML. Cancers (Basel) 2021; 13:cancers13143457. [PMID: 34298672 PMCID: PMC8304368 DOI: 10.3390/cancers13143457] [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: 06/16/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022] Open
Abstract
Nucleophosmin (NPM1) is a nucleocytoplasmic shuttling protein, predominantly located in the nucleolus, that regulates a multiplicity of different biological processes. NPM1 localization in the cell is finely tuned by specific signal motifs, with two tryptophan residues (Trp) being essential for the nucleolar localization. In acute myeloid leukemia (AML), several NPM1 mutations have been reported, all resulting in cytoplasmic delocalization, but the putative biological and clinical significance of different variants are still debated. We explored HOXA and HOXB gene expression profile in AML patients and found a differential expression between NPM1 mutations inducing the loss of two (A-like) Trp residues and those determining the loss of one Trp residue (non-A-like). We thus expressed NPM1 A-like- or non-A-like-mutated vectors in AML cell lines finding that NPM1 partially remained in the nucleolus in the non-A-like NPM1-mutated cells. As a result, only in A-like-mutated cells we detected HOXA5, HOXA10, and HOXB5 hyper-expression and p14ARF/p21/p53 pathway deregulation, leading to reduced sensitivity to the treatment with either chemotherapy or Venetoclax, as compared to non-A-like cells. Overall, we identified that the NPM1 mutational status mediates crucial biological characteristics of AML cells, providing the basis for further sub-classification and, potentially, management of this subgroup of patients.
Collapse
|
141
|
NPM1-mutated acute myeloid leukemia: from bench to bedside. Blood 2021; 136:1707-1721. [PMID: 32609823 DOI: 10.1182/blood.2019004226] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
The nucleophosmin (NPM1) gene encodes for a multifunctional protein with prominent nucleolar localization that shuttles between nucleus and cytoplasm. NPM1 mutations represent the most common genetic lesion in adult acute myeloid leukemia (AML; about one third of cases), and they act deterministically to cause the aberrant cytoplasmic delocalization of NPM1 mutants. Because of its unique features, NPM1-mutated AML is recognized as a distinct entity in the 2017 World Health Organization (WHO) classification of hematopoietic neoplasms. Here, we focus on recently identified functions of wild-type NPM1 in the nucleolus and address new biological and clinical issues related to NPM1-mutated AML. The relevance of the cooperation between NPM1 and other mutations in driving AML with different outcomes is presented. We also discuss the importance of eradicating NPM1-mutated clones to achieve AML cure and the impact of preleukemic clonal hematopoiesis persistence in predisposing to second AML. The contribution of HOX genes' expression to the development of NPM1-mutated AML is also highlighted. Clinically, yet unsolved diagnostic issues in the 2017 WHO classification of myeloid neoplasms and the importance of NPM1 mutations in defining the framework of European LeukemiaNet genetic-based risk stratification are discussed. Finally, we address the value and limits of NPM1-based measurable residual disease assessment for treatment guidance and present the results of promising preclinical studies with XPO1 and menin-MLL inhibitors.
Collapse
|
142
|
Guo C, Gao YY, Ju QQ, Zhang CX, Gong M, Li ZL. The landscape of gene co-expression modules correlating with prognostic genetic abnormalities in AML. J Transl Med 2021; 19:228. [PMID: 34051812 PMCID: PMC8164775 DOI: 10.1186/s12967-021-02914-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Background The heterogenous cytogenetic and molecular variations were harbored by AML patients, some of which are related with AML pathogenesis and clinical outcomes. We aimed to uncover the intrinsic expression profiles correlating with prognostic genetic abnormalities by WGCNA. Methods We downloaded the clinical and expression dataset from BeatAML, TCGA and GEO database. Using R (version 4.0.2) and ‘WGCNA’ package, the co-expression modules correlating with the ELN2017 prognostic markers were identified (R2 ≥ 0.4, p < 0.01). ORA detected the enriched pathways for the key co-expression modules. The patients in TCGA cohort were randomly assigned into the training set (50%) and testing set (50%). The LASSO penalized regression analysis was employed to build the prediction model, fitting OS to the expression level of hub genes by ‘glmnet’ package. Then the testing and 2 independent validation sets (GSE12417 and GSE37642) were used to validate the diagnostic utility and accuracy of the model. Results A total of 37 gene co-expression modules and 973 hub genes were identified for the BeatAML cohort. We found that 3 modules were significantly correlated with genetic markers (the ‘lightyellow’ module for NPM1 mutation, the ‘saddlebrown’ module for RUNX1 mutation, the ‘lightgreen’ module for TP53 mutation). ORA revealed that the ‘lightyellow’ module was mainly enriched in DNA-binding transcription factor activity and activation of HOX genes. The ‘saddlebrown’ module was enriched in immune response process. And the ‘lightgreen’ module was predominantly enriched in mitosis cell cycle process. The LASSO- regression analysis identified 6 genes (NFKB2, NEK9, HOXA7, APRC5L, FAM30A and LOC105371592) with non-zero coefficients. The risk score generated from the 6-gene model, was associated with ELN2017 risk stratification, relapsed disease, and prior MDS history. The 5-year AUC for the model was 0.822 and 0.824 in the training and testing sets, respectively. Moreover, the diagnostic utility of the model was robust when it was employed in 2 validation sets (5-year AUC 0.743–0.79). Conclusions We established the co-expression network signature correlated with the ELN2017 recommended prognostic genetic abnormalities in AML. The 6-gene prediction model for AML survival was developed and validated by multiple datasets. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02914-2.
Collapse
Affiliation(s)
- Chao Guo
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Ya-Yue Gao
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Qian-Qian Ju
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Chun-Xia Zhang
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Ming Gong
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China
| | - Zhen-Ling Li
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China.
| |
Collapse
|
143
|
How I diagnose and treat NPM1-mutated AML. Blood 2021; 137:589-599. [PMID: 33171486 DOI: 10.1182/blood.2020008211] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Mutations of the nucleophosmin (NPM1) gene, encoding for a nucleolar multifunctional protein, occur in approximately one-third of adult acute myeloid leukemia (AML). NPM1-mutated AML exhibits unique molecular, pathological, and clinical features, which led to its recognition as distinct entity in the 2017 World Health Organization (WHO) classification of myeloid neoplasms. Although WHO criteria for the diagnosis of NPM1-mutated AML are well established, its distinction from other AML entities may be difficult. Moreover, the percentage of blasts required to diagnose NPM1-mutated AML remains controversial. According to the European LeukemiaNet (ELN), determining the mutational status of NPM1 (together with FLT3) is mandatory for accurate relapse-risk assessment. NPM1 mutations are ideal targets for measurable residual disease (MRD) monitoring, since they are AML specific, frequent, very stable at relapse, and do not drive clonal hematopoiesis of undetermined significance. MRD monitoring by quantitative polymerase chain reaction of NPM1-mutant transcripts, possibly combined with ELN genetic-based risk stratification, can guide therapeutic decisions after remission. Furthermore, immunohistochemistry can be very useful in selected situations, such as diagnosis of NPM1-mutated myeloid sarcoma. Herein, we present 4 illustrative cases of NPM1-mutated AML that address important issues surrounding the biology, diagnosis, and therapy of this common form of leukemia.
Collapse
|
144
|
Retinoids in hematology: a timely revival? Blood 2021; 137:2429-2437. [PMID: 33651885 DOI: 10.1182/blood.2020010100] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/17/2021] [Indexed: 12/27/2022] Open
Abstract
The retinoic acid receptors (RARA, RARB, and RARG) are ligand-regulated nuclear receptors that act as transcriptional switches. These master genes drew significant interest in the 1990s because of their key roles in embryogenesis and involvement in a rare malignancy, acute promyelocytic leukemia (APL), in which the RARA (and very rarely, RARG or RARB) genes are rearranged, underscoring the central role of deregulated retinoid signaling in leukemogenesis. Several recent provocative observations have revived interest in the roles of retinoids in non-APL acute myeloid leukemia (AML), as well as in normal hematopoietic differentiation. We review the role of retinoids in hematopoiesis, as well as in the treatment of non-APL AMLs. From this perspective, broader uses of retinoids in the management of hematopoietic tumors are discussed.
Collapse
|
145
|
Exploiting Clonal Evolution to Improve the Diagnosis and Treatment Efficacy Prediction in Pediatric AML. Cancers (Basel) 2021; 13:cancers13091995. [PMID: 33919131 PMCID: PMC8122278 DOI: 10.3390/cancers13091995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 01/18/2023] Open
Abstract
Despite improvements in therapeutic protocols and in risk stratification, acute myeloid leukemia (AML) remains the leading cause of childhood leukemic mortality. Indeed, the overall survival accounts for ~70% but still ~30% of pediatric patients experience relapse, with poor response to conventional chemotherapy. Thus, there is an urgent need to improve diagnosis and treatment efficacy prediction in the context of this disease. Nowadays, in the era of high throughput techniques, AML has emerged as an extremely heterogeneous disease from a genetic point of view. Different subclones characterized by specific molecular profiles display different degrees of susceptibility to conventional treatments. In this review, we describe in detail this genetic heterogeneity of pediatric AML and how it is linked to relapse in terms of clonal evolution. We highlight some innovative tools to characterize minor subclones that could help to enhance diagnosis and a preclinical model suitable for drugs screening. The final ambition of research is represented by targeted therapy, which could improve the prognosis of pediatric AML patients, as well as to limit the side toxicity of current treatments.
Collapse
|
146
|
Kishtagari A, Levine RL. The Role of Somatic Mutations in Acute Myeloid Leukemia Pathogenesis. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a034975. [PMID: 32398288 DOI: 10.1101/cshperspect.a034975] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by attenuation of lineage differentiation trajectories that results in impaired hematopoiesis and enhanced self-renewal. To date, sequencing studies have provided a rich landscape of information on the somatic mutations that contribute to AML pathogenesis. These studies show that most AML genomes harbor relatively fewer mutations, which are acquired in a stepwise manner. Our understanding of the genetic basis of leukemogenesis informs a broader understanding of what initiates and maintains the AML clone and informs the development of prognostic models and mechanism-based therapeutic strategies. Here, we explore the current knowledge of genetic and epigenetic aberrations in AML pathogenesis and how recent studies are expanding our knowledge of leukemogenesis and using this to accelerate therapeutic development for AML patients.
Collapse
Affiliation(s)
- Ashwin Kishtagari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA.,Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
147
|
Wang E, Zhou H, Nadorp B, Cayanan G, Chen X, Yeaton AH, Nomikou S, Witkowski MT, Narang S, Kloetgen A, Thandapani P, Ravn-Boess N, Tsirigos A, Aifantis I. Surface antigen-guided CRISPR screens identify regulators of myeloid leukemia differentiation. Cell Stem Cell 2021; 28:718-731.e6. [PMID: 33450187 PMCID: PMC8145876 DOI: 10.1016/j.stem.2020.12.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 10/19/2020] [Accepted: 12/09/2020] [Indexed: 12/16/2022]
Abstract
Lack of cellular differentiation is a hallmark of many human cancers, including acute myeloid leukemia (AML). Strategies to overcome such a differentiation blockade are an approach for treating AML. To identify targets for differentiation-based therapies, we applied an integrated cell surface-based CRISPR platform to assess genes involved in maintaining the undifferentiated state of leukemia cells. Here we identify the RNA-binding protein ZFP36L2 as a critical regulator of AML maintenance and differentiation. Mechanistically, ZFP36L2 interacts with the 3' untranslated region of key myeloid maturation genes, including the ZFP36 paralogs, to promote their mRNA degradation and suppress terminal myeloid cell differentiation. Genetic inhibition of ZFP36L2 restores the mRNA stability of these targeted transcripts and ultimately triggers myeloid differentiation in leukemia cells. Epigenome profiling of several individuals with primary AML revealed enhancer modules near ZFP36L2 that associated with distinct AML cell states, establishing a coordinated epigenetic and post-transcriptional mechanism that shapes leukemic differentiation.
Collapse
Affiliation(s)
- Eric Wang
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA.
| | - Hua Zhou
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY 10016, USA
| | - Bettina Nadorp
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Geraldine Cayanan
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Xufeng Chen
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Anna H Yeaton
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Sofia Nomikou
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Matthew T Witkowski
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Sonali Narang
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Andreas Kloetgen
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Palaniraja Thandapani
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Niklas Ravn-Boess
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Aristotelis Tsirigos
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY 10016, USA; Institute for Computational Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Iannis Aifantis
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
148
|
HOXBLINC long non-coding RNA activation promotes leukemogenesis in NPM1-mutant acute myeloid leukemia. Nat Commun 2021; 12:1956. [PMID: 33782403 PMCID: PMC8007823 DOI: 10.1038/s41467-021-22095-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
Nucleophosmin (NPM1) is the most commonly mutated gene in acute myeloid leukemia (AML) resulting in aberrant cytoplasmic translocation of the encoded nucleolar protein (NPM1c+). NPM1c+ maintains a unique leukemic gene expression program, characterized by activation of HOXA/B clusters and MEIS1 oncogene to facilitate leukemogenesis. However, the mechanisms by which NPM1c+ controls such gene expression patterns to promote leukemogenesis remain largely unknown. Here, we show that the activation of HOXBLINC, a HOXB locus-associated long non-coding RNA (lncRNA), is a critical downstream mediator of NPM1c+-associated leukemic transcription program and leukemogenesis. HOXBLINC loss attenuates NPM1c+-driven leukemogenesis by rectifying the signature of NPM1c+ leukemic transcription programs. Furthermore, overexpression of HoxBlinc (HoxBlincTg) in mice enhances HSC self-renewal and expands myelopoiesis, leading to the development of AML-like disease, reminiscent of the phenotypes seen in the Npm1 mutant knock-in (Npm1c/+) mice. HoxBlincTg and Npm1c/+ HSPCs share significantly overlapped transcriptome and chromatin structure. Mechanistically, HoxBlinc binds to the promoter regions of NPM1c+ signature genes to control their activation in HoxBlincTg HSPCs, via MLL1 recruitment and promoter H3K4me3 modification. Our study reveals that HOXBLINC lncRNA activation plays an essential oncogenic role in NPM1c+ leukemia. HOXBLINC and its partner MLL1 are potential therapeutic targets for NPM1c+ AML. Nucleophosmin (NPM1) gene mutation induces a specific gene expression program leading to acute myeloid leukaemia. Here, the authors show that mutant NPM1 activates a HOXB locus-associated long non-coding RNA which is essential for its associated oncogenic transcriptional program and leukaemia development.
Collapse
|
149
|
Pei J, Wang G, Feng L, Zhang J, Jiang T, Sun Q, Ouyang L. Targeting Lysosomal Degradation Pathways: New Strategies and Techniques for Drug Discovery. J Med Chem 2021; 64:3493-3507. [PMID: 33764774 DOI: 10.1021/acs.jmedchem.0c01689] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A series of tools for targeted protein degradation are inspiring scientists to develop new drugs with advantages over traditional small-molecule drugs. Among these tools, proteolysis-targeting chimeras (PROTACs) are most representative of the technology based on proteasomes. However, the proteasome has little degradation effect on certain macromolecular proteins or aggregates, extracellular proteins, and organelles, which limits the application of PROTACs. Additionally, lysosomes play an important role in protein degradation. Therefore, lysosome-induced protein degradation drugs can directly regulate protein levels in vivo, achieve the goal of treating diseases, and provide new strategies for drug discovery. Lysosome-based degradation technology has the potential for clinical translation. In this review, strategies targeting lysosomal pathways and lysosome-based degradation techniques are summarized. In addition, lysosome-based degrading drugs are described, and the advantages and challenges are listed. Our efforts will certainly promote the design, discovery, and clinical application of drugs associated with this technology.
Collapse
Affiliation(s)
- Junping Pei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Lu Feng
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Tingting Jiang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
150
|
Grieselhuber NR, Mims AS. Novel Targeted Therapeutics in Acute Myeloid Leukemia: an Embarrassment of Riches. Curr Hematol Malig Rep 2021; 16:192-206. [PMID: 33738705 DOI: 10.1007/s11899-021-00621-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW Acute myeloid leukemia (AML) is an aggressive malignancy of the bone marrow that has a poor prognosis with traditional cytotoxic chemotherapy, especially in elderly patients. In recent years, small molecule inhibitors targeting AML-associated IDH1, IDH2, and FLT3 mutations have been FDA approved. However, the majority of AML cases do not have a targetable mutation. A variety of novel agents targeting both previously untargetable mutations and general pathways in AML are currently being investigated. Herein, we review selected new targeted therapies currently in early-phase clinical investigation in AML. RECENT FINDINGS The DOT1L inhibitor pinometostat in KMT2A-rearranged AML, the menin inhibitors KO-539 and SYNDX-5613 in KMT2Ar and NPM1-mutated AML, and the mutant TP53 inhibitor APR-246 are examples of novel agents targeting specific mutations in AML. In addition, BET inhibitors, polo-like kinase inhibitors, and MDM2 inhibitors are promising new drug classes for AML which do not depend on the presence of a particular mutation. AML remains in incurable disease for many patients but advances in genomics, epigenetics, and drug discovery have led to the development of many potential novel therapeutic agents, many of which are being investigated in ongoing clinical trials. Additional studies will be necessary to determine how best to incorporate these novel agents into routine clinical treatment of AML.
Collapse
Affiliation(s)
- Nicole R Grieselhuber
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Alice S Mims
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|