1
|
Sun H, Liu X, Wang L, Cui B, Mu W, Xia Y, Liu S, Liu X, Jiao Y, Zhao Y. Dexamethasone Sensitizes Acute Monocytic Leukemia Cells to Ara-C by Upregulating FKBP51. Front Oncol 2022; 12:888695. [PMID: 35860568 PMCID: PMC9290766 DOI: 10.3389/fonc.2022.888695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/09/2022] [Indexed: 11/26/2022] Open
Abstract
In this study, we demonstrated that the expression of FK506 binding protein 51 (FKBP51) is upregulated in acute monocytic leukemia (AML-M5) cells by dexamethasone and aimed to investigate the possible effects of FKBP51 on the growth and cytarabine sensitivity of AML-M5 cells. THP-1 and U937cells were used to establish AML-M5 cell models with FKBP51 overexpression and knockdown, respectively. Cell proliferation, apoptosis and response to cytarabine were investigated by cell cycle, CCK-8 and Flow cytometry analyses. The mice experiment was conducted to detect the role of FKBP51 on AML-M5 cells proliferation and antileukemia effect of Ara-C/Dexamethasone co-therapy in vivo. Western blots were employed to determine protein expression levels. FKBP51 upregulation significantly attenuated THP-1 cell proliferation and sensitized the cells to cytarabine treatment which was further enhanced by dexamethasone. These effects were indicated by decreases in cell viability, S-G2/M phase cell cycle distribution, cytarabine 50% inhibitory concentration (IC50) values and increases in apoptosis and were supported by decreased phosphorylation levels of AKT, GSK3β and FOXO1A and decreased levels of BCL-2 and increased levels of P21 and P27. In contrast, FKBP51 knockdown led to excessive U937 cell proliferation and cytarabine resistance, as indicated by increased cell viability and S-G2/M phase cell cycle distribution, decreased apoptosis, increased phosphorylation levels of AKT, GSK3β and FOXO1A, and increased BCL-2 and decreased P21 and P27 expression. In addition, an AKT inhibitor blocked cell cycle progression and reduced cell viability in all groups of cells. Furthermore, SAFit2, a specific FKBP51 inhibitor, increased U937 cell viability and cytarabine resistance as well as AKT phosphorylation. In conclusion, FKBP51 decelerates proliferation and improves the cytarabine sensitivity of AML-M5 cells by inhibiting AKT pathways, and dexamethasone in combination with Ara-C improves the chemosensitivity of AML-M5.
Collapse
Affiliation(s)
- Huanxin Sun
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xiaowen Liu
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Laicheng Wang
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Bin Cui
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Wenli Mu
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Yu Xia
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Shuang Liu
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xin Liu
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Yulian Jiao
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
- *Correspondence: Yulian Jiao, ; Yueran Zhao,
| | - Yueran Zhao
- Department of Central Laboratory, Shandong Provincial Hospital, Shandong University, Jinan, China
- Center for Reproductive Medicine, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
- *Correspondence: Yulian Jiao, ; Yueran Zhao,
| |
Collapse
|
2
|
Ramos DFV, Mancuso RI, Contieri B, Duarte A, Paiva L, de Melo Carrilho J, Saad STO, Lazarini M. Rac GTPases in acute myeloid leukemia cells: Expression profile and biological effects of pharmacological inhibition. Toxicol Appl Pharmacol 2022; 442:115990. [PMID: 35331739 DOI: 10.1016/j.taap.2022.115990] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/10/2022] [Accepted: 03/17/2022] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematological neoplasm with low survival rates. Thus, the investigation of new therapeutic targets is essential. The Rac subfamily of GTPase proteins has been shown to participate in the physiopathology of hematological malignancies. However, their expression and function in AML remain unclear. In this study, we evaluated Rac1, Rac2 and Rac3 gene expressions in AML and their impact on clinical outcomes. We further investigated the effects of the in vitro treatment with a Rac inhibitor (EHT-1864) on AML cell lines. Rac3 expression was increased in AML derived from myelodysplastic syndromes compared to healthy donors. Rac2 expression did not differ between AML patients and healthy donors, but de novo AML patients with higher Rac2 presented lower overall survival. Oncogenic pathway gene-sets related to AKT/mTOR were identified as associated with Rac1, Rac2 and Rac3 expressions. EHT-1864 treatment reduced the viability of OCI-AML3, KG1 and Kasumi-1 cells in a time and dose-dependent manner. In OCI-AML3 cells, treatment with EHT-1864 induced apoptosis, autophagy, and led to the accumulation of cells in the G1 phase of the cell cycle. These changes were concomitant with alterations in p53 and cyclins. Dowregulation of the PI3K/AKT/mTOR pathway was also observed. Interestingly, the combined treatment of EHT-1864 and low doses of daunorubicin enhanced OCI-AML3 cell apoptosis. In conclusion, Rac2 expression is a prognostic factor in AML and our preclinical results suggest that Rac inhibition may be an attractive mechanism to compose the antineoplastic strategy for this disease.
Collapse
Affiliation(s)
| | - Rubia Isler Mancuso
- Hematology and Bloood Transfusion Center, University of Campinas, Campinas, São Paulo, Brazil
| | - Bruna Contieri
- Department of Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Adriana Duarte
- Hematology and Bloood Transfusion Center, University of Campinas, Campinas, São Paulo, Brazil
| | - Luciana Paiva
- Hematology and Bloood Transfusion Center, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | - Mariana Lazarini
- Department of Pharmaceutical Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil; Hematology and Bloood Transfusion Center, University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
3
|
Role of the HOXA cluster in HSC emergence and blood cancer. Biochem Soc Trans 2021; 49:1817-1827. [PMID: 34374409 DOI: 10.1042/bst20210234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022]
Abstract
Hematopoiesis, the process of blood formation, is controlled by a complex developmental program that involves intrinsic and extrinsic regulators. Blood formation is critical to normal embryonic development and during embryogenesis distinct waves of hematopoiesis have been defined that represent the emergence of hematopoietic stem or progenitor cells. The Class I family of homeobox (HOX) genes are also critical for normal embryonic development, whereby mutations are associated with malformations and deformity. Recently, members of the HOXA cluster (comprising 11 genes and non-coding RNA elements) have been associated with the emergence and maintenance of long-term repopulating HSCs. Previous studies identified a gradient of HOXA expression from high in HSCs to low in circulating peripheral cells, indicating their importance in maintaining blood cell numbers and differentiation state. Indeed, dysregulation of HOXA genes either directly or by genetic lesions of upstream regulators correlates with a malignant phenotype. This review discusses the role of the HOXA cluster in both HSC emergence and blood cancer formation highlighting the need for further research to identify specific roles of these master regulators in normal and malignant hematopoiesis.
Collapse
|
4
|
Zhou H, Ning Y, Zeng G, Zhou C, Ding X. Curcumin promotes cell cycle arrest and apoptosis of acute myeloid leukemia cells by inactivating AKT. Oncol Rep 2021; 45:11. [PMID: 33649826 PMCID: PMC7877002 DOI: 10.3892/or.2021.7962] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/19/2021] [Indexed: 12/19/2022] Open
Abstract
Curcumin, a phytochemical from rhizomes of the plant Curcuma longa, has been reported to exert potential anticancer properties in various cancer types, including acute myeloid leukemia (AML). However, the underlying mechanism remains poorly understood. The present study demonstrated that curcumin had a stronger cytotoxic activity against AML cells compared with three other types of phytochemicals (epigallocatechin gallate, genistein and resveratrol). Protein phosphorylation profiling using an antibody array identified that curcumin treatment increased the phosphorylation levels of 14 proteins and decreased those of four proteins. A protein‑protein interaction network was constructed using the STRING database, in which AKT was identified as a hub protein with the highest connectivity (PRAS40, 4E‑BP1, P70S6K, RAF‑1 and p27). Western blotting results indicated that curcumin dose‑dependently suppressed the phosphorylation of AKT, PRAS40, 4E‑BP1, P70S6K, RAF‑1 and p27 in AML cell lines (ML‑2 and OCI‑AML5). It was also demonstrated that curcumin regulated the cell cycle‑ and apoptosis‑related proteins (cyclin D1, p21, Bcl2, cleaved‑caspase‑3 and cleaved‑PARP), leading to cell cycle arrest and apoptosis in both ML‑2 and OCI‑AML5 cells. These effects of curcumin were enhanced by the AKT inhibitor afuresertib but were suppressed by the AKT activator SC‑79, indicating that curcumin functions via AKT. In the AML xenograft mouse model, curcumin and afuresertib synergistically suppressed the engraftment, proliferation and survival of AML cells. Collectively, the present study demonstrated that curcumin exerted anti‑AML roles by inactivating AKT and these findings may aid in the treatment of AML.
Collapse
Affiliation(s)
- Hao Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yichong Ning
- Department of Clinical Laboratory, Chongzuo People's Hospital, Chongzuo, Guangxi 532200, P.R. China
| | - Guirong Zeng
- Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs and Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, Hunan 410331, P.R. China
| | - Chang Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiaofeng Ding
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| |
Collapse
|
5
|
A phase I study of a dual PI3-kinase/mTOR inhibitor BEZ235 in adult patients with relapsed or refractory acute leukemia. BMC Pharmacol Toxicol 2020; 21:70. [PMID: 32993794 PMCID: PMC7523358 DOI: 10.1186/s40360-020-00446-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 09/06/2020] [Indexed: 01/02/2023] Open
Abstract
Background Combined inhibition of phosphatidylinositol 3-kinase (PI3K) and the mammalian target of rapamycin (mTOR) complexes may be an efficient treatment for acute leukemia. The primary objective of this phase I single center open label study was to determine the maximum tolerated dose (MTD) and recommended phase II dose (RP2D) of the dual pan-class I PI3K and mTOR inhibitor BEZ235 in patients with advanced leukemia. Methods Herein patients > 18 years of age who had relapsed or showed refractory leukemia were treated with BEZ235 (orally at 300–400 mg BID (cohort − 1/1)) to assess safety, tolerability, preliminary efficacy and pharmacokinetic (PK). Adverse events data and serious adverse events were analyzed and haematological and clinical biochemistry toxicities were assessed from laboratory test parameters. Response was assessed for the first time at the end of cycle 1 (day 29) and after every subsequent cycle. Pharmacokinetic and pharmacodynamic analyses of BEZ235 were also included (BEZ235 plasma levels, phosphorylation of AKT, S6 and 4EBP1). On statistics this trial is a multiple ascending dose study in which a following variant of the 3 + 3 rule (“Rolling Six”), a minimum of 6 and a maximum of 12 patients was recruited for the dose escalation and another 5 were planned for the expansion phase. Results Twenty-four patients with ALL (n = 11) or AML (n = 12) or CML-BP (n = 1) were enrolled. All patients had failed one (n = 5) or more lines of therapy (n = 5) and 14 patients were in refractory / refractory relapse. No formal MTD was defined, stomatitis and gastrointestinal toxicity at 400 mg BID dose was considered incompatible with prolonged treatment. The RP2D of BEZ235 was defined as 300 mg BID. Four of 24 patients showed clinical benefit. Twenty-two of 24 patients discontinued because of progression, (median time to progression 27 days (4d-112d). There was no association between PK parameters and efficacy or tolerability. Conclusions Combined inhibition of PI3K and mTOR inhibits a clinically meaningful driver pathway in a small subset of patients with ALL, with no benefit in patients with AML. Trial registration ClinicalTrials.gov, identifier NCT01756118. retrospectively registered 19th December 2012, https://clinicaltrials.gov/ct2/show/NCT01756118.
Collapse
|
6
|
Guo C, Gao YY, Ju QQ, Zhang CX, Gong M, Li ZL. LINC00649 underexpression is an adverse prognostic marker in acute myeloid leukemia. BMC Cancer 2020; 20:841. [PMID: 32883226 PMCID: PMC7469387 DOI: 10.1186/s12885-020-07331-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/24/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNA) play a role in leukemogenesis, maintenance, development, and therapeutic resistance of AML. While few studies have focused on the prognostic significance of LINC00649 in AML, which we aim to investigate in this present study. METHODS We compared the expression level of LINC00649 between AML patients and healthy controls. The Kaplan-Meier curves of AML patients expressing high versus low level of LINC00649 was performed. The LINC00649 correlated genes/miRNAs/lncRNAs and methylation CpG sites were screened by Pearson correlation analysis with R (version 3.6.0), using TCGA-LAML database. The LINC00649 associated ceRNA network was established using lncBase 2.0 and miRWalk 2.0 online tools, combining results from correlation analysis. Finally, a prediction model was constructed using LASSO-Cox regression. RESULTS LINC00649 was underexpressed in bone marrow of AML group than that in healthy control group. The patients of LINC00649-low group have significantly inferior PFS and OS. A total of 154 mRNAs, 31 miRNAs, 28 lncRNAs and 1590 methylated CpG sites were identified to be significantly correlated with LINC00649. Furthermore, the network of ceRNA was established with 6 miRNAs and 122 mRNAs. The Lasso-Cox model fitted OS/PFS to novel prediction models, which integrated clinical factors, ELN risk stratification, mRNA/miRNA expression and methylation profiles. The analysis of time-dependent ROC for our model showed a superior AUC (AUC = 0.916 at 1 year, AUC = 0.916 at 3 years, and AUC = 0.891 at 5 years). CONCLUSIONS Low expression of LINC00649 is a potential unfavorable prognostic marker for AML patients, which requires the further validation. The analysis by LASSO-COX regression identified a novel comprehensive model with a superior diagnostic utility, which integrated clinical and genetic variables.
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
|
7
|
Guo C, Ju QQ, Zhang CX, Gong M, Li ZL, Gao YY. Overexpression of HOXA10 is associated with unfavorable prognosis of acute myeloid leukemia. BMC Cancer 2020; 20:586. [PMID: 32571260 PMCID: PMC7310421 DOI: 10.1186/s12885-020-07088-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
Background HOXA family genes were crucial transcription factors involving cell proliferation and apoptosis. While few studies have focused on HOXA10 in AML. We aimed to investigate the prognostic significance of HOXA10. Methods We downloaded datasets from GEO and BeatAML database, to compare HOXA expression level between AML patients and controls. Kaplan-Meier curves were used to estimate the impact of HOXA10 expression on AML survival. The differentially expressed genes, miRNAs, lncRNAs and methylated regions between HOXA10-high and -low groups were obtained using R (version 3.6.0). Accordingly, the gene set enrichment analysis (GSEA) was accomplished using MSigDB database. Moreover, the regulatory TFs/microRNAs/lncRNAs of HOXA10 were identified. A LASSO-Cox model fitted OS to clinical and HOXA10-associated genetic variables by glmnet package. Results HOXA10 was overexpressed in AML patients than that in controls. The HOXA10-high group is significantly associated with shorter OS and DFS. A total of 1219 DEGs, 131 DEmiRs, 282 DElncRs were identified to be associated with HOXA10. GSEA revealed that 12 suppressed and 3 activated pathways in HOXA10-high group. Furthermore, the integrated regulatory network targeting HOXA10 was established. The LASSO-Cox model fitted OS to AML-survival risk scores, which included age, race, molecular risk, expression of IKZF2/LINC00649/LINC00839/FENDRR and has-miR-424-5p. The time dependent ROC indicated a satisfying AUC (1-year AUC 0.839, 3-year AUC 0.871 and 5-year AUC 0.813). Conclusions Our study identified HOXA10 overexpression as an adverse prognostic factor for AML. The LASSO-COX regression analysis revealed novel prediction model of OS with superior diagnostic utility.
Collapse
Affiliation(s)
- Chao Guo
- 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
| | - Ya-Yue Gao
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, China.
| |
Collapse
|
8
|
Brattås MK, Reikvam H, Tvedt THA, Bruserud Ø. Precision medicine for TP53-mutated acute myeloid leukemia. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1644164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
| | - Håkon Reikvam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Øystein Bruserud
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Section for Hematology, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
9
|
Carneiro BA, Kaplan JB, Altman JK, Giles FJ, Platanias LC. Targeting mTOR signaling pathways and related negative feedback loops for the treatment of acute myeloid leukemia. Cancer Biol Ther 2015; 16:648-56. [PMID: 25801978 PMCID: PMC4622839 DOI: 10.1080/15384047.2015.1026510] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/01/2015] [Indexed: 12/29/2022] Open
Abstract
An accumulating understanding of the complex pathogenesis of acute myeloid leukemia (AML) continues to lead to promising therapeutic approaches. Among the key aberrant intracellular signaling pathways involved in AML, the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) axis is of major interest. This axis modulates a wide array of critical cellular functions, including proliferation, metabolism, and survival. Pharmacologic inhibitors of components of this pathway have been developed over the past decade, but none has an established role in the treatment of AML. This review will discuss the preclinical data and clinical results driving ongoing attempts to exploit the PI3K/AKT/mTOR pathway in patients with AML and address issues related to negative feedback loops that account for leukemic cell survival. Targeting the PI3K/AKT/mTOR pathway is of high interest for the treatment of AML, but combination therapies with other targeted agents may be needed to block negative feedback loops in leukemia cells.
Collapse
Affiliation(s)
- Benedito A Carneiro
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Jason B Kaplan
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Jessica K Altman
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Francis J Giles
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
| | - Leonidas C Platanias
- Robert H Lurie Comprehensive Cancer Center of Northwestern University; Chicago, IL, USA
- Division of Hematology and Oncology and Northwestern Medicine Developmental Therapeutics Institute; Northwestern University; Feinberg School of Medicine; Chicago, IL, USA
- Division of Hematology-Oncology; Department of Medicine; Jesse Brown VA Medical Center; Chicago, IL, USA
| |
Collapse
|